PMID-sentid Pub_year Sent_text comp_official_name comp_offset protein_name organism prot_offset 17696279-4 2007 Administration of vanadium compounds suppresses c-fos/c-jun expression and AP-1 activity, resulting in inhibition of MMP expression in response to factors such as interleukin 1 (IL-1). Vanadium 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 17696279-7 2007 NAC in combination with vanadate appeared to increase the efficacy of c-fos/c-jun inhibition, while decreasing toxicity. Acetylcysteine 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 17696279-7 2007 NAC in combination with vanadate appeared to increase the efficacy of c-fos/c-jun inhibition, while decreasing toxicity. Vanadates 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 17287821-4 2007 In order to elucidate the neuroanatomical substrates underlying the effect of the maximal effective dose of THC, we investigated cFos expression in anxiety-related brain regions (prefrontal cortex, nucleus accumbens, amygdala, and hippocampus) of rats exposed to the EPM. Dronabinol 108-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-133 17287821-5 2007 THC significantly lowered the amount of cFos in prefrontal cortex and amygdala without affecting the other cerebral areas. Dronabinol 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-44 17287824-9 2007 Nicotine induced c-fos expression in the bed nucleus of the stria terminalis, the central nucleus of the amygdala (CeA), nucleus accumbens, and the superior colliculus (SC) at both ages, whereas it activated the hypothalamic paraventricular nucleus (PVN) and consequent corticosterone secretion only in adults. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 17287824-10 2007 Acetaldehyde potentiated nicotine-induced c-fos in CeA and SC, and activation of PVN c-fos expression/plasma corticosterone release; however, this drug interaction was only observed in behaviorally tested animals, not those that were minimally stressed. Acetaldehyde 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 17287824-10 2007 Acetaldehyde potentiated nicotine-induced c-fos in CeA and SC, and activation of PVN c-fos expression/plasma corticosterone release; however, this drug interaction was only observed in behaviorally tested animals, not those that were minimally stressed. Nicotine 25-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 17763989-1 2007 An immunohistochemical method was used to study the expression of transcription factor c-Fos in the mid part of the main olfactory bulbs in 18-day-old rats after training to seek their mothers using an olfactory orientation cue (propionic acid) in a Y-maze. propionic acid 229-243 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 17763989-3 2007 The propionic acid odor evoked an insignificant increase in c-Fos expression, predominantly in the granular layer of the dorsomedial area of the olfactory bulb. propionic acid 4-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 17669374-0 2007 Brainstem areas activated by diazepam withdrawal as measured by Fos-protein immunoreactivity in rats. Diazepam 29-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 17703884-6 2007 Using Fos protein as a marker of neuronal activation, we found that systemic administration of PMX53 reduced the LPS-induced activation of paraventricular corticotropin-releasing factor (PVN CRF) and central amygdala cells. AcPhe(ornithine-Pro-cyclohexylamine-Trp-Arg) 95-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 6-9 17761543-9 2007 Both Devalue and Maintain rats showed greater FOS expression than control rats in amygdala central nucleus, GC, and both subregions of ACBs. acbs 135-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 17725832-10 2007 NGF treatment and intravesical saline distention (2 hr) increased expression of Fos protein in L6-S1 spinal cord and altered the distribution pattern of Fos-IR cells. Sodium Chloride 31-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 17725832-10 2007 NGF treatment and intravesical saline distention (2 hr) increased expression of Fos protein in L6-S1 spinal cord and altered the distribution pattern of Fos-IR cells. Sodium Chloride 31-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-156 17662531-0 2007 Suppression of spike-wave discharge activity and c-fos expression by 2-methyl-4-oxo-3H-quinazoline-3-acetyl piperidine (Q5) in vivo. 2-methyl-4-oxo-3H-quinazoline-3-acetylpiperidine 69-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 17553493-8 2007 Pre-treatment with dexamethasone reduced OT secretion, as well as Fos-OT immunoreactive neurons in response to both isotonic and hypertonic BVE. Dexamethasone 19-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 17393298-11 2007 Additionally, we also observed an increase in c-fos mRNA expression in both PVN and SON 6 h after water deprivation, which progressively decreased 24, 48, and 72 h after the onset of water deprivation. Water 98-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 17393298-11 2007 Additionally, we also observed an increase in c-fos mRNA expression in both PVN and SON 6 h after water deprivation, which progressively decreased 24, 48, and 72 h after the onset of water deprivation. Water 183-188 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 17442060-5 2007 In addition, ammonia affected Sp1 and c-fos, transcription factors that regulate CNTF mRNA and protein expression, which showed partial dephosphorylation and significantly lower mRNA and protein levels. Ammonia 13-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 17631915-0 2007 Induction of c-Fos and zif268 in the nociceptive amygdala parallel abstinence hyperalgesia in rats briefly exposed to morphine. Morphine 118-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 17631915-5 2007 Surprisingly, in many regions c-Fos induction by morphine was reduced or blocked by naloxone, even though these subjects had also experienced the effects of morphine for 30 min prior to antagonist administration. Morphine 49-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 17631915-5 2007 Surprisingly, in many regions c-Fos induction by morphine was reduced or blocked by naloxone, even though these subjects had also experienced the effects of morphine for 30 min prior to antagonist administration. Naloxone 84-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 17531342-0 2007 Expansion of formalin-evoked Fos-immunoreactivity in rats with a spinal cord injury. Formaldehyde 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 17531342-6 2007 Stereological analysis of the sham group revealed that the extent of formalin-induced Fos expression was within the lumbar dorsal horn, with numerous Fos-like immunoreactive profiles in the ipsilateral dorsal horn and some contralateral immunoreactive profiles. Formaldehyde 69-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 17531342-6 2007 Stereological analysis of the sham group revealed that the extent of formalin-induced Fos expression was within the lumbar dorsal horn, with numerous Fos-like immunoreactive profiles in the ipsilateral dorsal horn and some contralateral immunoreactive profiles. Formaldehyde 69-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-153 17582689-7 2007 administration of DOI affected Fos-like immunoreactivity (-LI) evoked by formalin injection into the masseter muscle. Formaldehyde 73-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 17582689-9 2007 Formalin-evoked Fos-LI was significantly reduced in laminae I-II of the Vc/C2, but not vl-Vi/Vc region after i.c.v. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 17495048-5 2007 Microinjections of the GABA(A) receptor antagonist, bicuculline (20 nl, 1 mm), into the PF region elicited cortical and hippocampal activation, increased the respiratory rate and hypoglossal nerve activity, induced c-fos expression in ORX and other PF neurones, and increased c-fos expression in pontine A7 and other noradrenergic neurones. Bicuculline 52-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 215-220 17714194-4 2007 The distribution of VTA-projecting neurons activated by amphetamine was examined by combining retrograde transport of the cholera toxin beta subunit (CTb), injected into the VTA, with immunodetection of Fos. Amphetamine 56-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 203-206 17714194-5 2007 The quantitative analysis of CTb-Fos double labelling demonstrates that amphetamine induced a rapid activation of Fos in a large number of brain areas projecting to the VTA. Amphetamine 72-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 17714194-5 2007 The quantitative analysis of CTb-Fos double labelling demonstrates that amphetamine induced a rapid activation of Fos in a large number of brain areas projecting to the VTA. Amphetamine 72-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 17562390-0 2007 Suppression by intrathecal BmK IT2 on rat spontaneous pain behaviors and spinal c-Fos expression induced by formalin. Formaldehyde 108-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 17562390-2 2007 It was found that the formalin-induced rat spontaneous flinches and spinal c-Fos expression could be significantly suppressed by intrathecal BmK IT2 pre- or post-formalin injection in a dose-dependent manner. Formaldehyde 22-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 17562390-2 2007 It was found that the formalin-induced rat spontaneous flinches and spinal c-Fos expression could be significantly suppressed by intrathecal BmK IT2 pre- or post-formalin injection in a dose-dependent manner. Formaldehyde 162-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 17562390-5 2007 In addition, the suppression by intrathecal BmK IT2 on formalin-induced c-Fos expression in superficial laminae was more significant than that in deeper laminae. Formaldehyde 55-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 17495048-5 2007 Microinjections of the GABA(A) receptor antagonist, bicuculline (20 nl, 1 mm), into the PF region elicited cortical and hippocampal activation, increased the respiratory rate and hypoglossal nerve activity, induced c-fos expression in ORX and other PF neurones, and increased c-fos expression in pontine A7 and other noradrenergic neurones. Bicuculline 52-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 276-281 17532793-0 2007 Identification of central nervous system sites involved in the water diuresis response elicited by central microinjection of nociceptin/ Orphanin FQ in conscious rats via c-Fos and inducible cAMP early repressor immunocytochemistry. Water 63-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-176 17549049-1 2007 BACKGROUND AND PURPOSE: We have previously demonstrated that morphine withdrawal induced hyperactivity of the heart by activation of noradrenergic pathways innervating the left and right ventricle, as evaluated by noradrenaline turnover and c-Fos expression. Morphine 61-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 241-246 17549049-7 2007 KEY RESULTS: Naloxone-induced morphine withdrawal activated ERK1/2 and increased c-Fos expression in cardiac tissues. Naloxone 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 17549049-7 2007 KEY RESULTS: Naloxone-induced morphine withdrawal activated ERK1/2 and increased c-Fos expression in cardiac tissues. Morphine 30-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 17430989-3 2007 We then evaluated EB effects on Isop-induced Fos immunoreactivity (Fos-IR) in the hindbrain baroreflex circuit. ethylbenzene 18-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 17430989-3 2007 We then evaluated EB effects on Isop-induced Fos immunoreactivity (Fos-IR) in the hindbrain baroreflex circuit. ethylbenzene 18-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 17543352-3 2007 In addition, the role of group I mGluRs in dorsal horn neuronal Fos expression was determined in tetrodotoxin (TTX)-treated in vitro spinal cords of naive rats and those with Complete Freund"s Adjuvant (CFA) peripheral inflammation. Tetrodotoxin 97-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 17543352-3 2007 In addition, the role of group I mGluRs in dorsal horn neuronal Fos expression was determined in tetrodotoxin (TTX)-treated in vitro spinal cords of naive rats and those with Complete Freund"s Adjuvant (CFA) peripheral inflammation. Tetrodotoxin 111-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 17543352-7 2007 In the dorsal horn of naive but not CFA rats, DHPG increased Fos expression and this was reduced by MPEP and both PKC and ERK inhibitors. 6-methyl-2-(phenylethynyl)pyridine 100-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 17356878-10 2007 Spinal administration of either YM872 or MK-801 reduced Fos expression in the spinal cord at all ages. YM 872 32-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 17543464-6 2007 Exposure to a mild stressor elevated an index of norepinephrine turnover in the prefrontal cortex and also increased Fos expression in tyrosine hydroxylase-positive LC neurons in rats exposed to prenatal cocaine but not in rats exposed to prenatal saline. Cocaine 204-211 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 17543464-6 2007 Exposure to a mild stressor elevated an index of norepinephrine turnover in the prefrontal cortex and also increased Fos expression in tyrosine hydroxylase-positive LC neurons in rats exposed to prenatal cocaine but not in rats exposed to prenatal saline. Sodium Chloride 248-254 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 17356878-10 2007 Spinal administration of either YM872 or MK-801 reduced Fos expression in the spinal cord at all ages. Dizocilpine Maleate 41-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 17350615-8 2007 Intracerebroventricular pretreatment with indomethacin (an inhibitor of cyclooxygenase) abolished the CRF- and AVP-induced cFos expression in all regions described above. Indomethacin 42-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-127 17350615-9 2007 On the other hand, intracerebroventricular pretreatment with furegrelate (an inhibitor of thromboxane A2 synthase) abolished the CRF-induced cFos expression in the adrenal A-cells, but not in the celiac ganglia, and also abolished the AVP-induced cFos expression in both A-cells and NA-cells in the adrenal medulla. furegrelate 61-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-145 17350615-9 2007 On the other hand, intracerebroventricular pretreatment with furegrelate (an inhibitor of thromboxane A2 synthase) abolished the CRF-induced cFos expression in the adrenal A-cells, but not in the celiac ganglia, and also abolished the AVP-induced cFos expression in both A-cells and NA-cells in the adrenal medulla. furegrelate 61-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 247-251 17436292-8 2007 Furthermore, the behavioral sequence of eating and increased activity exhibited by DE animals in the minutes following water drinking is accompanied by a further increase in the number of Fos-ir nuclei in the LHA. Water 119-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 188-191 17434543-11 2007 Fos-immunoreactivity increased in orexin (OX) neurons after insulin, from 8.7+/-4.9% after saline injection to 37+/-9% after insulin. Sodium Chloride 91-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 17442279-11 2007 Using immunohistochemical methods, brain c-Fos protein expression was analyzed in rats that extinguished the CTA as well as those that exhibited SR of the CTA after a 30-day latency. Chlorotrianisene 109-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 17459353-0 2007 AM404 decreases Fos-immunoreactivity in the spinal cord in a model of inflammatory pain. N-(4-hydroxyphenyl)arachidonylamide 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 17459353-5 2007 We found that Fos-positive neurons in dorsal superficial and deep laminae of the lumbar spinal cord increased in formalin-injected animals and that AM404 significantly reduced Fos induction. Formaldehyde 113-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 17459353-5 2007 We found that Fos-positive neurons in dorsal superficial and deep laminae of the lumbar spinal cord increased in formalin-injected animals and that AM404 significantly reduced Fos induction. N-(4-hydroxyphenyl)arachidonylamide 148-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 176-179 17694681-0 2007 Effect of intrathecal NG-nitro-L-arginine methyl ester administration on Fos expression in the spinal dorsal horn in rats following sciatic nerve ligation. NG-Nitroarginine Methyl Ester 22-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-76 17694681-3 2007 In this study, we examined the effect of intrathecal L-NAME, a non-selective nitric oxide synthase (NOS) inhibitor, on nociceptive behavior and spinal Fos expression in rats following chronic constriction injury (CCI) of sciatic nerve, a model of neuropathic pain similar to that observed in clinical setting. NG-Nitroarginine Methyl Ester 53-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-154 17694681-7 2007 RESULTS: As compared with untreated animals, both CCI and sham operations evoked an early and long-term Fos expression, whereas a significant decrease in PWL was demonstrated only in rats receiving CCI. CCI 50-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 17525596-9 2007 RESULTS: Although the rats subjected to neonatal peripheral carrageenan injection developed mechanical hypoalgesia in bilateral hind paws at baseline, they displayed increased spinal cord Fos expression at 2 h and exaggerated mechanical pain hypersensitivity at 4 h (but not at other time points) after plantar incision. Carrageenan 60-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 188-191 17522523-10 2007 Furthermore, CPZ reduced the percentage of double-labeled neurons for Fos and TH in the RVLM of the rat brain. capsazepine 13-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 17524563-16 2007 Pre-treatment with dexamethasone induced a significant decrease in Fos immunoreactivity in these nuclei compared with the vehicle. Dexamethasone 19-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 17462630-0 2007 Central neural distribution of immunoreactive Fos and CRH in relation to plasma ACTH and corticosterone during sepsis in the rat. Corticosterone 89-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 17462630-6 2007 The number of Fos-positive cell nuclei in the NTS on D3 and D4 did not differ but had greater variance on D3 than on D4 (P<0.01) with a divergent response in the PM of D3 that was correlated with plasma ACTH (r=0.927, P<0.01) but not with corticosterone. Corticosterone 245-259 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 17382304-0 2007 Effect of the selective dopamine D3 receptor antagonist SB-277011-A on regional c-Fos-like expression in rat forebrain. SB 277011 56-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 17382304-2 2007 We employed c-Fos immunohistochemistry to characterise the functional neuroanatomical effects of acute administration of SB-277011-A and observed a time-dependent increase in the density of c-Fos-like positive nuclei in rat forebrain with maximal effects observed 2 h post-dose. SB 277011 121-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 17382304-2 2007 We employed c-Fos immunohistochemistry to characterise the functional neuroanatomical effects of acute administration of SB-277011-A and observed a time-dependent increase in the density of c-Fos-like positive nuclei in rat forebrain with maximal effects observed 2 h post-dose. SB 277011 121-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 190-195 17420096-0 2007 Acute nicotine enhances c-fos mRNA expression differentially in reward-related substrates of adolescent and adult rat brain. Nicotine 6-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 17420096-2 2007 To help determine the potential brain circuitry involved, we investigated the effect of acute nicotine administration (0.4 or 0.8mg/kg, s.c.) on the expression of c-fos mRNA in the brains of adolescent (P35) and adult (P67-70) male Wistar rats using in situ hybridization. Nicotine 94-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-168 17420096-3 2007 Nicotine administration increased c-fos mRNA expression in several brain regions, including the central amygdala, locus coeruleus, nucleus accumbens core, paraventricular nucleus of the hypothalamus and lateral septum of adolescent and adult rats. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 17420096-4 2007 Nicotine increased c-fos mRNA expression more robustly in the bed nucleus of the stria terminalis, nucleus accumbens shell and ventral tegmental area in adolescent rats. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 17360123-4 2007 ABA renewal of alcohol-seeking was associated with selective increases in c-Fos protein induction in basolateral amygdala, ventral accumbens shell, and lateral hypothalamus (renewal-associated Fos). alisol B 23-acetate 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 17360123-4 2007 ABA renewal of alcohol-seeking was associated with selective increases in c-Fos protein induction in basolateral amygdala, ventral accumbens shell, and lateral hypothalamus (renewal-associated Fos). alisol B 23-acetate 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 17360123-4 2007 ABA renewal of alcohol-seeking was associated with selective increases in c-Fos protein induction in basolateral amygdala, ventral accumbens shell, and lateral hypothalamus (renewal-associated Fos). Alcohols 15-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 17360123-4 2007 ABA renewal of alcohol-seeking was associated with selective increases in c-Fos protein induction in basolateral amygdala, ventral accumbens shell, and lateral hypothalamus (renewal-associated Fos). Alcohols 15-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 17360123-8 2007 However, c-Fos induction in either lateral hypothalamic orexin-negative or orexin-positive neurons was positively and significantly correlated with alcohol-seeking. Alcohols 148-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 17493276-6 2007 We further demonstrate that intrathecal injection of SP-CTA in rats induces the phosphorylation of the transcription factor cyclic AMP response element binding protein (CREB) and also enhances the expression of the immediate early gene c-Fos. sp-cta 53-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 236-241 17336288-0 2007 Effects of the fatty acid amide hydrolase inhibitor URB597 on the sleep-wake cycle, c-Fos expression and dopamine levels of the rat. cyclohexyl carbamic acid 3'-carbamoylbiphenyl-3-yl ester 52-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 17336288-8 2007 We additionally found out that, compared to controls, c-Fos immunoreactivity in hypothalamus and dorsal raphe nucleus was increased in rats that received URB597, oleoylethanolamide or palmitoylethanolamide (10, 20 microg/5 microl, i.c.v.). cyclohexyl carbamic acid 3'-carbamoylbiphenyl-3-yl ester 154-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 17336288-8 2007 We additionally found out that, compared to controls, c-Fos immunoreactivity in hypothalamus and dorsal raphe nucleus was increased in rats that received URB597, oleoylethanolamide or palmitoylethanolamide (10, 20 microg/5 microl, i.c.v.). oleoylethanolamide 162-180 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 17537436-6 2007 Fos expression was elevated in AVP-positive magnocellular PVN neurons and AVP- and corticotropin releasing hormone (CRH)-positive parvocellular PVN neurons after water restriction. Water 162-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 17336288-8 2007 We additionally found out that, compared to controls, c-Fos immunoreactivity in hypothalamus and dorsal raphe nucleus was increased in rats that received URB597, oleoylethanolamide or palmitoylethanolamide (10, 20 microg/5 microl, i.c.v.). palmidrol 184-205 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 17426598-4 2007 Water avoidance stimulated fecal pellet output, which was associated with Fos expression in myenteric ganglia of proximal and distal colon including in a population of peripheral choline acetyltransferase-immunoreactive neurons. Water 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 17544397-10 2007 Compared to sham-depleted animals there was a significantly greater number of Fos-/FG-ir double-labeled cells in the subfornical organ, the organum vasculosum of the lamina terminalis and the median preoptic nucleus in rats that ingested NaCl. Sodium Chloride 238-242 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 17418497-9 2007 Both SSP-sap and SP-sap reduced formalin-induced FOS expression in deep and superficial laminae of the L4 dorsal horn in parallel with the reduction in phase 2 behavior. ssp-sap 5-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 17418497-9 2007 Both SSP-sap and SP-sap reduced formalin-induced FOS expression in deep and superficial laminae of the L4 dorsal horn in parallel with the reduction in phase 2 behavior. Formaldehyde 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 17443818-0 2007 Differential effects of stress and amphetamine administration on Fos-like protein expression in corticotropin releasing factor-neurons of the rat brain. Amphetamine 35-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 17443818-8 2007 The present results indicate that stress and amphetamine elicited a distinct pattern of brain Fos-like protein expression and differentially activated some of the brain CRF neuronal populations, despite similar levels of overall FLI in the case of IMO and amphetamine. Amphetamine 45-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 17234710-8 2007 Ablation of vagal afferent function by perivagal application of capsaicin, a specific afferent neurotoxin, abolished c-fos protein immunoreactivity, suggesting that activation of the NTS due to GLP-2 is dependent on vagal afferents. Capsaicin 64-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 16872852-0 2007 Hypertonic saline-induced muscle nociception and c-fos activation are partially mediated by peripheral NMDA receptors. Sodium Chloride 11-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 17433545-0 2007 Expression of c-Fos protein in the brain after intravenous injection of ghrelin in rats. Ghrelin 72-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 17433545-1 2007 In this study, we surveyed central neurons that might be activated after peripheral administration of a gut-brain peptide ghrelin, by examining neurons expressing c-Fos protein. Ghrelin 122-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-168 17433545-4 2007 Secondly, we examined central neurons that expressed c-Fos protein after intravenous injection of ghrelin. Ghrelin 98-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 17433545-5 2007 We found that intravenously injected ghrelin induced the neural expression of c-Fos protein in several nuclei and circumventricular organs in the brain. Ghrelin 37-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 17333448-14 2007 Adding indigestible sugars, such as FOS or live microbial as L. casei, in the diet significantly improves daidzein protective effects on the skeleton. Sugars 20-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-39 17304580-9 2007 Parturition-induced Fos-positive neurons in the spinal cords were abundant in control rats, but were reduced by 70% in MPEP-treated animals. 6-methyl-2-(phenylethynyl)pyridine 119-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 17433371-4 2007 The trypsin-induced spinal Fos expression was completely abolished by oral pre-administration of camostat mesilate at 300 mg/kg. camostat 97-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 17333448-14 2007 Adding indigestible sugars, such as FOS or live microbial as L. casei, in the diet significantly improves daidzein protective effects on the skeleton. daidzein 106-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-39 21179752-9 2007 (4) EGb, Que, Captopril and DPI all decreased Ang II-stimulated early response gent c-fos mnRNA expression. Quercetin 9-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 17654838-3 2007 On the other hand, it was of particular interest in the experiment of the AP that strong expression of gamma aminobutylic acid (GABA) frequently showed similar pattern of distribution to that of c-Fos in the PAG. gamma aminobutylic acid 103-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-200 17654838-3 2007 On the other hand, it was of particular interest in the experiment of the AP that strong expression of gamma aminobutylic acid (GABA) frequently showed similar pattern of distribution to that of c-Fos in the PAG. gamma-Aminobutyric Acid 128-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-200 21179752-9 2007 (4) EGb, Que, Captopril and DPI all decreased Ang II-stimulated early response gent c-fos mnRNA expression. Captopril 14-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 21179752-9 2007 (4) EGb, Que, Captopril and DPI all decreased Ang II-stimulated early response gent c-fos mnRNA expression. 3-aminodiphenyleneiodium 28-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 17425902-6 2007 Conversely, c-Fos expression in the basolateral and medial amygdala was lower in the COND group as compared to the UNCD group. uncd 115-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 21179752-9 2007 (4) EGb, Que, Captopril and DPI all decreased Ang II-stimulated early response gent c-fos mnRNA expression. gent 79-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 17425902-7 2007 Furthermore, c-Fos expression was particularly high in the medial amygdala of the UNCD group. uncd 82-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 17098214-5 2007 METHODS: Rats were tested in a conditioned reinstatement model of relapse with subsequent examination of brain c-fos expression patterns elicited by an EtOH S(+) versus a cue associated with nonreward (S(-)). Ethanol 152-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 21179752-10 2007 CONCLUSION: EGb and Que could inhibit AngII-induced cardiomyocyte hypertrophy through a ROS-dependent pathway, the effect of Que might be related to the JNK and c-fos cascade. Quercetin 20-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 17098214-7 2007 RESULTS: The EtOH S(+) reinstated extinguished responding and increased c-fos expression within the prefrontal cortex, hippocampus, nucleus accumbens, and hypothalamic paraventricular nucleus (PVN). Ethanol 13-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 17129577-8 2007 In addition, cFos immunoreactivity studies show H(3) antagonists activate neuronal cells in restricted rat brain regions in contrast to widespread activation after modafinil or amphetamine treatment. HS 3 48-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-17 17098214-8 2007 Naltrexone suppressed the S(+)-induced reinstatement and attenuated hippocampal CA3 c-fos expression, while increasing neural activity in the extended amygdala and PVN. Naltrexone 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 17288999-0 2007 Effects of acute heat exposure on prosencephalic c-Fos expression in normohydrated, water-deprived and salt-loaded rats. Water 84-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 17316993-0 2007 Fos-like immunoreactivity in rat dorsal raphe nuclei induced by alkaloid extract of Mitragyna speciosa. Alkaloids 64-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 17514764-0 2007 c-fos gene expression is increased in the paraventricular hypothalamic nucleus of Sprague-Dawley rats with visceral pain induced by acetic acid without detectable changes of corticotrophin-releasing factor mRNA: a quantitative approach with an image analysis system. Acetic Acid 132-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 17337280-5 2007 Cyrneine A enhanced activation of the transcription factors activator protein-1 (AP-1) and nuclear factor-kappaB, but not CREB, and this was accompanied by enhanced c-fos expression. cyrneine A 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 17514764-2 2007 The c-fos gene expression in the paraventricular hypothalamic nucleus (PVN) of Sprague-Dawley rats for an animal model for visceral or somatovisceral pain induced by 2% acetic acid (AA) was used in this study. Acetic Acid 169-180 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 17624190-9 2007 Cadmium chloride at the doses of 5, 10, and 20 micromol/kg induced expression of proto-oncogenes c-myc, c-fos, and c-jun. Cadmium Chloride 0-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 17194726-8 2007 Fos-ir in the forebrain CVOs, the subfornical organ, and organum vasculosum laminae terminalis was consistently elevated only by hyperosmotic NaCl. Sodium Chloride 142-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 17194726-10 2007 Hyperosmotic NaCl greatly elevated Fos-ir in the area postrema, but even glucose and urea caused moderate elevations that may be related to volume expansion rather than osmolality. Sodium Chloride 13-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 17624190-14 2007 CONCLUSION: Cadmium at 5-20 micromol/kg can induce hepatocellular DNA damage, expression of proto-oncogenes c-myc, c-fos, and c-jun as well as apoptosis in rats. Cadmium 12-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 17194737-6 2007 Moreover, after intracerebroventricular N-PCT administration, Fos protein, a marker of neuronal activation, was found in regions of primary importance in the integration of hormonal signals for energy homeostasis and feeding. n-pct 40-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 18225445-7 2007 The hypothalamic consequences of glucose infusion were studied by c-Fos protein immunodetection. Glucose 33-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 17255209-11 2007 Expression of Fos was increased in the area postrema and the nucleus of the tractus solitarius by using immunohistochemistry after icv and iv injection of ghrelin. Ghrelin 155-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 17355316-11 2007 Analyses of brains from saline-treated VGX animals revealed a significant induction of Fos in the nucleus tractus solitarii and changes in agouti-related peptide and pro-opiomelanocortin mRNA expression in the hypothalamus compared to their SHAM counterparts, indicating that the vagotomy surgery itself induced a modification of brain activity in areas involved in regulating appetite. Sodium Chloride 24-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 17204558-10 2007 Moreover, sucrose attenuated restraint-induced c-fos mRNA expression in the basolateral amygdala, infralimbic cortex, and claustrum. Sucrose 10-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 17278128-3 2007 Strikingly, we found densely Fos-labeled neurons in all divisions of the MGB after both presentation of an auditory stimulus and administration of a gamma-aminobutyric acid type A (GABA(A)) antagonist (bicuculline methobromide; BIM) to the auditory cortex. gamma-Aminobutyric Acid 149-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 17278128-3 2007 Strikingly, we found densely Fos-labeled neurons in all divisions of the MGB after both presentation of an auditory stimulus and administration of a gamma-aminobutyric acid type A (GABA(A)) antagonist (bicuculline methobromide; BIM) to the auditory cortex. gamma-Aminobutyric Acid 181-185 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 17278128-3 2007 Strikingly, we found densely Fos-labeled neurons in all divisions of the MGB after both presentation of an auditory stimulus and administration of a gamma-aminobutyric acid type A (GABA(A)) antagonist (bicuculline methobromide; BIM) to the auditory cortex. bicuculline methobromide 202-226 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 17278128-6 2007 The dorsal (DCIC) and external cortices (ECIC) of the IC ipsilateral to the BIM-injected cortex showed a significantly higher number of Fos-labeled neurons than the contralateral IC. bim 76-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-139 17063151-2 2007 As there is a dense projection of orexin neurons from the lateral hypothalamus to A10 dopaminergic neurons, and some antipsychotics have been shown to increase the expression of c-fos in orexin-containing cells in the hypothalamus, we hypothesized that stimulation of orexin receptors plays a role in the effects of antipsychotics on the activity of A9 and A10 dopamine cells. Dopamine 86-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 178-183 17190973-0 2007 Long-lasting reductions of ethanol drinking, enhanced ethanol-induced sedation, and decreased c-fos expression in the Edinger-Westphal nucleus in Wistar rats exposed to the organophosphate chlorpyrifos. organophosphate chlorpyrifos 173-201 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 17190973-7 2007 An immunocytochemical assay revealed reduced c-fos expression in the Edinger-Westphal nucleus following CPF treatment, a critical brain area that has been implicated in ethanol intake and sedation. Ethanol 169-176 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 17092780-0 2007 Brain Fos expression during 48 h after cisplatin treatment: neural pathways for acute and delayed visceral sickness. Cisplatin 39-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 6-9 17224133-4 2007 RESULTS: Chronic paroxetine treatment reduced the immobility scores during forced swimming, confirming the previously observed antidepressant-like effect in these animals, and attenuated the forced swim-induced c-Fos response in a restricted set (11 of 70) of brain areas. Paroxetine 17-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 211-216 17289273-3 2007 The present study investigated whether ambient temperature influences MDMA-induced expression of Fos, a marker of neural activation. N-Methyl-3,4-methylenedioxyamphetamine 70-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 17289273-6 2007 However MDMA-induced Fos expression was augmented in 15 of 30 brain regions at the high temperature. N-Methyl-3,4-methylenedioxyamphetamine 8-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 17289273-8 2007 MDMA-induced Fos expression was unaffected by ambient temperature at many other sites, and was diminished at high temperature at one site (the islands of Calleja), suggesting that the effect of temperature on MDMA-induced Fos expression was not a general pharmacokinetic effect. N-Methyl-3,4-methylenedioxyamphetamine 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 17289273-8 2007 MDMA-induced Fos expression was unaffected by ambient temperature at many other sites, and was diminished at high temperature at one site (the islands of Calleja), suggesting that the effect of temperature on MDMA-induced Fos expression was not a general pharmacokinetic effect. N-Methyl-3,4-methylenedioxyamphetamine 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 222-225 17289273-8 2007 MDMA-induced Fos expression was unaffected by ambient temperature at many other sites, and was diminished at high temperature at one site (the islands of Calleja), suggesting that the effect of temperature on MDMA-induced Fos expression was not a general pharmacokinetic effect. N-Methyl-3,4-methylenedioxyamphetamine 209-213 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 222-225 17207573-0 2007 Fluoxetine attenuates the effects of pentylenetetrazol on rat freezing behavior and c-Fos expression in the dorsomedial periaqueductal gray. Fluoxetine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 17207573-4 2007 Simultaneously, PTZ significantly increased c-Fos expression in the dorsomedial periaqueductal gray (DMPAG), examined 2h after the retention trial, in comparison to the control group (p<0.01). Pentylenetetrazole 16-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 17207573-4 2007 Simultaneously, PTZ significantly increased c-Fos expression in the dorsomedial periaqueductal gray (DMPAG), examined 2h after the retention trial, in comparison to the control group (p<0.01). dmpag 101-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 17226797-3 2007 A subset of neurons within the nucleus tractus solitarius (NTS) shows c-Fos activation during prolonged sodium deprivation in rats. Sodium 104-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 17204372-2 2007 To determine whether these patterns are present during early postnatal life, and whether they change during development, Fos expression was assessed following acute exposure to single aliphatic acid odors in developing rats beginning at postnatal day 3 (P3). Fatty Acids 184-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-124 17082349-0 2007 Progressive postnatal increases in Fos immunoreactivity in the forebrain and brain stem of rats after viscerosensory stimulation with lithium chloride. Lithium Chloride 134-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 17082349-5 2007 Compared with saline treatment, LiCl increased Fos only slightly in the area postrema, nucleus of the solitary tract, and lateral parabrachial nucleus on P0. Lithium Chloride 32-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 17082349-7 2007 Maximal Fos responses to LiCl were observed on P14 in all areas except the BNST, in which LiCl-induced Fos activation continued to increase through P28. Lithium Chloride 25-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-11 17082349-7 2007 Maximal Fos responses to LiCl were observed on P14 in all areas except the BNST, in which LiCl-induced Fos activation continued to increase through P28. Lithium Chloride 25-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-106 17082349-7 2007 Maximal Fos responses to LiCl were observed on P14 in all areas except the BNST, in which LiCl-induced Fos activation continued to increase through P28. Lithium Chloride 90-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-11 17082349-7 2007 Maximal Fos responses to LiCl were observed on P14 in all areas except the BNST, in which LiCl-induced Fos activation continued to increase through P28. Lithium Chloride 90-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-106 17082349-8 2007 These results indicate that central LiCl-sensitive interoceptive circuits in rats are not fully functional at birth, and show age-dependent increases in neural Fos responses to viscerosensory stimulation with LiCl. Lithium Chloride 36-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-163 17082349-8 2007 These results indicate that central LiCl-sensitive interoceptive circuits in rats are not fully functional at birth, and show age-dependent increases in neural Fos responses to viscerosensory stimulation with LiCl. Lithium Chloride 209-213 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-163 17082351-7 2007 In addition, Fos-LI in the DVC in response to both exogenous CCK-8 and camostat administration was significantly attenuated by vagotomy, as well as by blocking CCK(1) receptors. camostat 71-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 17136530-0 2007 Modulation of neuronal activity in CNS pain pathways following propofol administration in rats: Fos and EEG analysis. Propofol 63-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-99 17136530-1 2007 We studied Fos expression in the central nociceptive pathways at different sedative levels in order to clarify the central mechanism of propofol"s nociceptive action. Propofol 136-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 17136530-9 2007 Fos expression was significantly greater in the Vc and Periaqueductal gray following greater amount of PRO infusions compared, whereas they were significantly smaller in the Vc in the rats with PEN infusion. Pentobarbital 194-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 17430619-0 2007 Nociception after intraperitoneal injection of a sodium pentobarbitone formulation with and without lidocaine in rats quantified by expression of neuronal c-fos in the spinal cord--a preliminary study. Pentobarbital 49-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-160 17430619-2 2007 In the present pilot study in rats, spinal nociception after intraperitoneal injection of sodium pentobarbitone, with and without lidocaine, was examined by estimation of the number of c-fos-expressing neurones in the spinal dorsal horn. Pentobarbital 90-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 185-190 17409705-7 2007 The c-Fos immunohistochemical study indicated that the medial prefrontal cortex is an action site of imipramine in ACTH-treated rats. Imipramine 101-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 17276011-0 2007 Fos and glutamate AMPA receptor subunit coexpression associated with cue-elicited cocaine-seeking behavior in abstinent rats. Cocaine 82-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 17276011-2 2007 We have reported an increase in neuronal activation in rats, measured by Fos protein expression, in various limbic and cortical regions following exposure to cocaine-associated cues. Cocaine 158-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-76 17204251-0 2007 C-fos expression in the rat brain following lithium chloride-induced illness. Lithium Chloride 44-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 17204251-1 2007 The present study examined c-Fos expression in selected brain areas consequent to administration of lithium chloride, the typical illness-inducing agent used in laboratory studies of conditioned taste aversion. Lithium Chloride 100-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 17204251-3 2007 New findings indicate significant lithium-induced c-Fos in the gustatory region of the thalamus and the bed nucleus of the stria terminalis but not in the insular cortex. Lithium 34-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 17068161-9 2007 Twelve hours after PEG was injected in rats that had been sodium deprived for 4 days, the HSD2 neurons showed a consistent increase in c-Fos immunoreactivity. Polyethylene Glycols 19-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 17110529-9 2007 Ondansetron (1 mg/kg) significantly attenuated duodenal intralipid-induced Fos-LI in the dorsal hindbrain. Ondansetron 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 17110529-9 2007 Ondansetron (1 mg/kg) significantly attenuated duodenal intralipid-induced Fos-LI in the dorsal hindbrain. soybean oil, phospholipid emulsion 56-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 17327451-9 2007 gAd induced mRNA expression of c-fos and c-jun and activated extracellular signal-regulated kinase. ganoderic acid D 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 17214984-0 2007 Increased dietary sodium alters Fos expression in the lamina terminalis during intravenous angiotensin II infusion. Sodium 18-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 17214984-1 2007 These studies examined the effects of increased dietary sodium on expression of Fos, the protein product of c-fos, in forebrain structures in the rat following intravenous infusion with angiotensin II (AngII). Sodium 56-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 17120244-9 2007 In luciferase reporter assays, DRE, which actively represses c-fos by binding the calcium-binding transcriptional repressor DREAM, was activated by glutamate, whereas SRE and CRE were not. Calcium 82-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 17214984-7 2007 Furthermore, Fos-Li IR in the MnPO was increased following AngII infusion in rats consuming a high sodium diet, but not in animals drinking Tap. Sodium 99-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 17169339-6 2007 In contrast, combination of both dopamine agonists produced a greater increase than SKF38393 did in the mRNA levels of c-fos in the nucleus accumbens, striatum and substantia nigra. Dopamine 33-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 17523441-10 2007 At the same time, ketanserin administration led to a significant decrease in the level of c-Fos protein in the hypothalamus in OVX rats as compared to the intact control. Ketanserin 18-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 17294041-8 2007 An immunohistochemical SABC assay was used to describe the expression profiles of c-Fos-positive cells in different brain regions including the CeA, hippocampus, cortex, hypothalamus and periaqueductal gray (PAG) at 1, 2, 4 and 8 h after bilateral single-administration of PBG by i.c.a. phenyl biguanide 273-276 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 17161392-1 2007 We previously reported that brief (1 h), but not extended (6 h), daily access to cocaine results in a sensitized locomotor response to cocaine and in elevated c-Fos immunoreactivity and DAT binding in the nucleus accumbens (N.Acc) core. Cocaine 81-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-164 17120244-0 2007 Glutamate activates c-fos in glial cells via a novel mechanism involving the glutamate receptor subtype mGlu5 and the transcriptional repressor DREAM. Glutamic Acid 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 17120244-2 2007 Glutamate activates c-fos in neurons through the calcium-dependent phosphorylation of CREB by ERK and/or CaMKIV kinase pathways downstream NMDA-receptors. Glutamic Acid 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 17120244-2 2007 Glutamate activates c-fos in neurons through the calcium-dependent phosphorylation of CREB by ERK and/or CaMKIV kinase pathways downstream NMDA-receptors. Calcium 49-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 17120244-3 2007 In glial cells, however, the activation of c-fos by glutamate is poorly understood. Glutamic Acid 52-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 17120244-4 2007 Because glial cells actively modulate neuronal excitability and the brain"s response to injury, we studied the mechanisms by which glutamate activates c-fos in rat cortical glial cells. Glutamic Acid 131-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-156 17120244-5 2007 Glutamate potently induced c-fos mRNA in a calcium-dependent manner, as demonstrated by using the calcium chelator BAPTA-AM. Glutamic Acid 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 17120244-5 2007 Glutamate potently induced c-fos mRNA in a calcium-dependent manner, as demonstrated by using the calcium chelator BAPTA-AM. Calcium 43-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 17120244-5 2007 Glutamate potently induced c-fos mRNA in a calcium-dependent manner, as demonstrated by using the calcium chelator BAPTA-AM. Calcium 98-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 17120244-5 2007 Glutamate potently induced c-fos mRNA in a calcium-dependent manner, as demonstrated by using the calcium chelator BAPTA-AM. 1,2-bis(2-aminophenoxy)ethane N,N,N',N'-tetraacetic acid acetoxymethyl ester 115-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 17120244-6 2007 Glutamate-induced c-fos mRNA expression was not sensitive to inhibitors of ERK, p38(MAPK), or CaMK pathways, indicating that glial c-fos is activated by a distinct mechanism. Glutamic Acid 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 17120244-6 2007 Glutamate-induced c-fos mRNA expression was not sensitive to inhibitors of ERK, p38(MAPK), or CaMK pathways, indicating that glial c-fos is activated by a distinct mechanism. Glutamic Acid 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-136 17120244-7 2007 Thapsigargin abolished the glutamate effect on c-fos mRNA, indicating ER calcium mobilization. Thapsigargin 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 17120244-7 2007 Thapsigargin abolished the glutamate effect on c-fos mRNA, indicating ER calcium mobilization. Glutamic Acid 27-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 17120244-7 2007 Thapsigargin abolished the glutamate effect on c-fos mRNA, indicating ER calcium mobilization. Calcium 73-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 17120244-8 2007 Additionally, glutamate induction of c-fos mRNA was sensitive to the mGluR5 antagonist MPEP but not the NMDA-R antagonist MK-801. Glutamic Acid 14-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 17120244-8 2007 Additionally, glutamate induction of c-fos mRNA was sensitive to the mGluR5 antagonist MPEP but not the NMDA-R antagonist MK-801. 6-methyl-2-(phenylethynyl)pyridine 87-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 17173980-5 2007 CLP led to increased plasma nitrate levels, protein leakage and hypotension and caused mortality of 80% by 24 h. Expression of c-fos in paraventricular (PVN), supraoptic (SON) and organum vasculosum of lamina terminalis (OVLT) nuclei, as well as plasma AVP concentration were increased at 6 h but reduced to basal levels 24 h after CLP. Nitrates 28-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 17173980-6 2007 Aminoguanidine pre-treatment prevented the increase in plasma nitrate levels and hypotension in the first 6 h. It also reduced AVP secretion and hypothalamic c-fos expression. pimagedine 0-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 17120244-9 2007 In luciferase reporter assays, DRE, which actively represses c-fos by binding the calcium-binding transcriptional repressor DREAM, was activated by glutamate, whereas SRE and CRE were not. Glutamic Acid 148-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 17120244-10 2007 Finally, glutamate caused the nuclear export of DREAM in astrocytes, and transfection of astrocytes with a mutant variant of DREAM that constitutively binds DNA inhibited glutamate-induced c-Fos expression. Glutamic Acid 9-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 189-194 17120244-10 2007 Finally, glutamate caused the nuclear export of DREAM in astrocytes, and transfection of astrocytes with a mutant variant of DREAM that constitutively binds DNA inhibited glutamate-induced c-Fos expression. Glutamic Acid 171-180 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 189-194 17120244-11 2007 These findings are in sharp contrast to the mechanism described in neurons and suggest a novel pathway activated by glutamate in glial cells that employs mGluR5, ER calcium, and the derepression of c-fos at the DRE. Glutamic Acid 116-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 198-203 17123639-4 2007 In contrast, Fos expression in stress-associated brain areas, including the ventral lateral bed nucleus of the stria terminalis (VL-BNST), central nucleus of the amygdala (CE), and noradrenergic (A2) neurons in the nucleus tractus solitarius (NTS) was significantly elevated only in morphine-abstinent animals. Morphine 283-291 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 16990492-4 2007 However, c-Fos-like immunoreactivity was markedly attenuated in these brain regions when chylomicron formation/secretion was blocked by Pluronic L-81. l-81 145-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 16990492-7 2007 The present study also revealed that c-Fos-positive neurons induced by feeding of Intalipid were abolished by CCK type 1 receptor antagonist, Lorglumide. intalipid 82-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 16990492-7 2007 The present study also revealed that c-Fos-positive neurons induced by feeding of Intalipid were abolished by CCK type 1 receptor antagonist, Lorglumide. lorglumide 142-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 17622784-1 2007 AIM: To investigate the relationships between c-fos expression and oxidative stress or inflammation in the liver induced by acute cadmium (Cd) exposure. Cadmium 130-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 17622784-1 2007 AIM: To investigate the relationships between c-fos expression and oxidative stress or inflammation in the liver induced by acute cadmium (Cd) exposure. Cadmium 139-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 17622784-6 2007 RESULTS: Acute Cd exposure significantly increased MDA levels, decreased GSH levels and upregulated the gene expression of TNF-alpha and c-fos in the liver. Cadmium 15-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 17622784-9 2007 CONCLUSION: Our data suggested that c-fos induction is independent of oxidative stress or inflammation in the liver during the process of acute Cd exposure in rats. Cadmium 144-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 17202671-10 2007 Pre-treatment of VSMCs with NQ304 (1-10 microM) was found to significantly inhibit the 5% FBS-induced phosphorylations of ERK1/2 and Akt, the activation of AP-1 and the expression of c-fos. vsmcs 17-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 183-188 17202671-10 2007 Pre-treatment of VSMCs with NQ304 (1-10 microM) was found to significantly inhibit the 5% FBS-induced phosphorylations of ERK1/2 and Akt, the activation of AP-1 and the expression of c-fos. NQ304 28-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 183-188 17216288-1 2007 We previously demonstrated that morphine withdrawal induced hyperactivity of the heart by the activation of noradrenergic pathways innervating the left and right ventricle, as evaluated by noradrenaline (NA) turnover and Fos expression. Morphine 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 221-224 17084866-8 2007 The 5-HT(3) receptor is only partially implicated in quipazine-induced expression of c-FOS, while NMDA receptor inhibition blocks quipazine photic-like effects on both parameters. Quipazine 53-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 16850320-8 2007 While mean numbers of GR-plus Fos-ir neurons in the PVH, DMH, LHA, and NTS, but not the PVT were significantly elevated after one dose of NPH, this increase was abolished in each site by RIIH. Dimenhydrinate 57-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 16897600-0 2007 Kynurenine in combination with probenecid mitigates the stimulation-induced increase of c-fos immunoreactivity of the rat caudal trigeminal nucleus in an experimental migraine model. Kynurenine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 16897600-0 2007 Kynurenine in combination with probenecid mitigates the stimulation-induced increase of c-fos immunoreactivity of the rat caudal trigeminal nucleus in an experimental migraine model. Probenecid 31-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 16897600-1 2007 Nitroglycerin, often used as a migraine model, results in increased number of c-fos immunoreactive secondary sensory neurons in the caudal trigeminal nucleus. Nitroglycerin 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 16897600-4 2007 Systemic kynurenine + probenecid treatment significantly diminishes nitroglycerin-induced increase of c-fos immunoreactivity in the brainstem. Kynurenine 9-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 16897600-4 2007 Systemic kynurenine + probenecid treatment significantly diminishes nitroglycerin-induced increase of c-fos immunoreactivity in the brainstem. Probenecid 22-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 16897600-4 2007 Systemic kynurenine + probenecid treatment significantly diminishes nitroglycerin-induced increase of c-fos immunoreactivity in the brainstem. Nitroglycerin 68-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 17028028-1 2007 Acute administration of clozapine has been reported to activate the locus coeruleus (LC) and beta-adrenoceptor-dependent Fos immunoreactivity in the medial prefrontal cortex (mPFC) in rodents. Clozapine 24-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-124 17028028-2 2007 Haloperidol is reported to exhibit a similar acute effect on LC firing and beta-adrenoceptor dependent Fos immunoreactivity in the mPFC but only at high doses. Haloperidol 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-106 17284934-10 2007 c-Fos protein was detected in the corneal epithelium around the area of ethanol exposure from 60 to 120 min after the treatment, while c-Jun protein was not detected. Ethanol 72-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 17027755-1 2007 This study examined the effects of dehydration and rehydration with water on Fos and FosB staining in the brainstem of rats. Water 68-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-80 17873287-0 2007 Gabapentin completely attenuated the acute morphine induced c-Fos expression in the rat striatum. Gabapentin 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 17873287-0 2007 Gabapentin completely attenuated the acute morphine induced c-Fos expression in the rat striatum. Morphine 43-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 17873287-2 2007 Therefore, we examined the combined effects of GBP-Morphine on acute morphine induced c-Fos expression in rat striatum. Morphine 69-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 17873287-13 2007 Present results showed that GBP-morphine combination action prevented the acute morphine induced c-Fos expression in rat striatum. Morphine 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 17873287-13 2007 Present results showed that GBP-morphine combination action prevented the acute morphine induced c-Fos expression in rat striatum. Morphine 80-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 17873368-0 2007 Effect of gabapentin on c-Fos expression in the CNS after paw surgery in rats. Gabapentin 10-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 17012605-4 2007 Glibenclamide, a nonselective K(ATP) channel blocker reversed the antihypertrophic effect of all three AR agonists as determined by cell size and atrial natriuretic peptide expression and early c-fos up-regulation. Glyburide 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 194-199 17703089-5 2007 While the rats fed NCM supplemented with glucose or fat displayed an equally strong amylin-induced activation as fasted rats or rats fed plain NCM, a significantly lower c-Fos expression was observed in rats fed a protein-supplemented NCM or a NCM containing all three nutrients. Glucose 41-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-175 17216288-6 2007 Moreover, morphine withdrawal induces Fos expression, an enhancement of NA turnover and an increase in the total TH levels. Morphine 10-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 16875883-8 2007 These findings suggest that the inhibitory effects of NQ304 on DNA synthesis, proliferation, and cell cycle progression on PDGF-BB-stimulated VSMCs are mediated via the downregulations of AP-1 activation and c-fos expression achieved in turn via the suppressions of the phosphatidylinositol 3-kinase (PI3K)/Akt and ERK1/2 signaling pathways. NQ304 54-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 208-213 16962079-8 2006 Although cocaine increased levels of c-fos mRNA, none of the estrogen or progesterone replacement paradigms affected this measure. Cocaine 9-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 16935424-8 2006 Naloxone challenge to morphine-treated animals precipitated an intense withdrawal syndrome that depleted CGRP-immunoreactivity and increased Fos expression in the dorsal horn. Naloxone 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 16935424-8 2006 Naloxone challenge to morphine-treated animals precipitated an intense withdrawal syndrome that depleted CGRP-immunoreactivity and increased Fos expression in the dorsal horn. Morphine 22-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 16935424-12 2006 Treatment with AM-251 also reduced the depletion of CGRP, suppressed Fos-induction, and prevented the increase in capsaicin-evoked spinal CGRP release. AM 251 15-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 17113947-0 2006 Zolpidem, a selective GABA(A) receptor alpha1 subunit agonist, induces comparable Fos expression in oxytocinergic neurons of the hypothalamic paraventricular and accessory but not supraoptic nuclei in the rat. Zolpidem 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 17113947-4 2006 Zolpidem elicited a concordant Fos/OXY staining in all four PVN sub-areas investigated, including the anterior (15.71+/-2.35%), middle (14.52+/-2.53%), dorsal (13.34+/-2.61%), and periventricular (18.21+/-4.75%) ones, however, had no significant stimulatory effect on OXY cells in the SON. Zolpidem 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 17122581-9 2006 Furthermore, by inhibiting the ERK pathway, propofol blocked NMDA receptor-dependent activation of two key transcription factors, Elk-1 and cyclic adenosine monophosphate response element-binding protein (CREB), and, as a result, attenuated Elk-1/CREB-dependent reporter gene (c-Fos) expression. Propofol 44-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 277-282 16962193-0 2006 Effects on c-Fos expression in the PAG and thalamus by selective input via tetrodotoxin-resistant afferent fibres from muscle and skin. Tetrodotoxin 75-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 17184865-5 2007 CBX decreased the number of Fos/NeuN-IR neurons (25+/-1.7), but did not affect Fos/GFAP-IR astrocytes (16.2+/-5.4), compared with vehicle-preadministered rats (Fos/NeuN-IR neurons 135+/-4.2, and Fos/GFAP-IR astrocytes 25.8+/-4). Carbenoxolone 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 16979827-6 2006 In order to assess the effect of antagonizing 5-HT2A or 5-HT3 receptors on formalin-induced Fos-LI, rats were pre-treated with local (masseter muscle) administration of ketanserin or tropisetron (0.01, 0.1 mg/rat) 20 min prior to formalin injection. Formaldehyde 75-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 16949214-9 2006 SCH23390 also suppressed basal and renewal-associated c-Fos protein induction throughout accumbens, and, selectively suppressed renewal-associated c-Fos induction in lateral hypothalamus. SCH 23390 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 16949214-9 2006 SCH23390 also suppressed basal and renewal-associated c-Fos protein induction throughout accumbens, and, selectively suppressed renewal-associated c-Fos induction in lateral hypothalamus. SCH 23390 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-152 17026529-0 2006 PACAP and C2-ceramide generate different AP-1 complexes through a MAP-kinase-dependent pathway: involvement of c-Fos in PACAP-induced Bcl-2 expression. N-acetylsphingosine 10-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 17026529-4 2006 Thus, PACAP increased the proportion of c-Fos and Jun D while C2-ceramide increased c-Jun and reduced c-Fos in AP-1 complexes. N-acetylsphingosine 62-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 17026529-6 2006 The effect of PACAP on c-Fos expression was blocked by the mitogen-activated protein kinase/extracellular signal-regulated kinase (MEK) inhibitor, U0126, while phosphorylation of c-Jun induced by C2-ceramide was abrogated by the protein phosphatase 2A (PP2A) inhibitor, okadaic acid. U 0126 147-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 16855532-9 2006 Forty-three genes exhibited significant differences in expression in HR vs LR 24 h after METH treatment including a group of immediate-early genes (IEGs) (eg, c-fos, junB, NGFI-B, serum-regulated glucocorticoid kinase). Methamphetamine 89-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-164 17270257-5 2007 In males, but not in females, there were effects of nicotine treatment and of stress condition on Fos immunoreactive (Fos-ir) cell counts in the paraventricular nucleus (PVN) of the hypothalamus: SS males had higher Fos-ir counts than did ISO and non-stressed control males, and higher Fos-ir counts in the PVN were found in repeated-nicotine groups than in acute-nicotine and saline groups. Nicotine 52-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 17270257-5 2007 In males, but not in females, there were effects of nicotine treatment and of stress condition on Fos immunoreactive (Fos-ir) cell counts in the paraventricular nucleus (PVN) of the hypothalamus: SS males had higher Fos-ir counts than did ISO and non-stressed control males, and higher Fos-ir counts in the PVN were found in repeated-nicotine groups than in acute-nicotine and saline groups. Nicotine 52-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 17270257-5 2007 In males, but not in females, there were effects of nicotine treatment and of stress condition on Fos immunoreactive (Fos-ir) cell counts in the paraventricular nucleus (PVN) of the hypothalamus: SS males had higher Fos-ir counts than did ISO and non-stressed control males, and higher Fos-ir counts in the PVN were found in repeated-nicotine groups than in acute-nicotine and saline groups. Nicotine 52-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 17270257-5 2007 In males, but not in females, there were effects of nicotine treatment and of stress condition on Fos immunoreactive (Fos-ir) cell counts in the paraventricular nucleus (PVN) of the hypothalamus: SS males had higher Fos-ir counts than did ISO and non-stressed control males, and higher Fos-ir counts in the PVN were found in repeated-nicotine groups than in acute-nicotine and saline groups. Nicotine 52-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 16395306-4 2006 Rats trained on a running wheel under the influence of cocaine for 4 days subsequently displayed greater c-fos induction by cocaine than untrained controls. Cocaine 55-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 16395306-4 2006 Rats trained on a running wheel under the influence of cocaine for 4 days subsequently displayed greater c-fos induction by cocaine than untrained controls. Cocaine 124-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 16395306-7 2006 Wheel training was performed after injection of cocaine (25 mg/kg) or vehicle, and c-fos induction by a cocaine challenge was measured 24 h later. Cocaine 104-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 16395306-8 2006 Rats that trained under cocaine (but not vehicle) showed a greater c-fos response in the striatum compared to locked-wheel controls. Cocaine 24-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 16982036-8 2006 Glutathione (GSH) depletion produced by TMMC activated extracellular signal-regulated kinase (ERK), which led to c-Fos expression and transactivation of activator protein 1 (AP-1) and HO-1 gene expression in the HSCs. Glutathione 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 16982036-8 2006 Glutathione (GSH) depletion produced by TMMC activated extracellular signal-regulated kinase (ERK), which led to c-Fos expression and transactivation of activator protein 1 (AP-1) and HO-1 gene expression in the HSCs. Glutathione 13-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 16982036-8 2006 Glutathione (GSH) depletion produced by TMMC activated extracellular signal-regulated kinase (ERK), which led to c-Fos expression and transactivation of activator protein 1 (AP-1) and HO-1 gene expression in the HSCs. 2',4',6'-tris(methoxymethoxy) chalcone 40-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 16901937-4 2006 In the presence of 0.8 mM Ca(2+), very low O(2) medium induced an increase in c-fos expression throughout the VMS. o(2) medium 43-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 16901937-5 2006 The reduction of synaptic transmission and blockade of the respiratory drive by 0.2 mM Ca(2+)-1.6 mM Mg(2+) abolished c-fos expression in the medial VMS (at the lateral edge of the pyramidal tract) but not in the perifacial retrotrapezoid nucleus/parafacial respiratory group (RTN/pFRG) VMS, suggesting the existence of perifacial RTN/pFRG hypoxia-sensing neurons. magnesium ion 101-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 17047458-7 2006 Pretreatment with melatonin also reduced the number of Fos and nitric oxide synthase immunoreactive cells in the trigeminal nucleus caudalis. Melatonin 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 17006656-0 2006 Fos immunoreactivity in some locomotor neural centres of 6OHDA-lesioned rats. Oxidopamine 57-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 17006656-7 2006 In conclusion, we reveal an increase in the number of strongly labelled Fos+ cells within the cuneiform nucleus of the so-called defensive locomotive system in 6OHDA-lesioned rats. Oxidopamine 160-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 16962193-3 2006 Here, we investigated the c-Fos expression in the thalamus and the periaqueductal grey matter (PAG) induced by electrical stimulation of tetrodotoxin-resistant (TTX-r), presumably nociceptive, afferent fibres. pag 95-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 16962193-3 2006 Here, we investigated the c-Fos expression in the thalamus and the periaqueductal grey matter (PAG) induced by electrical stimulation of tetrodotoxin-resistant (TTX-r), presumably nociceptive, afferent fibres. Tetrodotoxin 137-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 16962193-3 2006 Here, we investigated the c-Fos expression in the thalamus and the periaqueductal grey matter (PAG) induced by electrical stimulation of tetrodotoxin-resistant (TTX-r), presumably nociceptive, afferent fibres. ttx-r 161-166 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 16962193-7 2006 c-Fos was visualized using DAB immunohistochemistry. diazobenzenesulfonic acid 27-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 16962193-8 2006 Here we report for the first time that in the PAG and medial thalamus, the main effect of TTX-r input from muscle was a reduction in c-Fos expression, and that in some thalamic nuclei (e.g. posterior, reuniens, and central medial nuclei), significant differences in the number of c-Fos-positive cells were found after muscle and cutaneous input, respectively. ttx-r 90-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 16962193-8 2006 Here we report for the first time that in the PAG and medial thalamus, the main effect of TTX-r input from muscle was a reduction in c-Fos expression, and that in some thalamic nuclei (e.g. posterior, reuniens, and central medial nuclei), significant differences in the number of c-Fos-positive cells were found after muscle and cutaneous input, respectively. ttx-r 90-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 280-285 16997409-10 2006 Icv administration of CP-472555 during antecedent hypoglycemia prevented RIIH-associated reductions in Fos expression by these neurons. CP 472555 22-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-106 17385422-4 2006 The increase of c-Fos-positive cells number in 2 hours after LPS injection was observed in AFTN, PVH, LHA, VMH, DMH and PH. Dimenhydrinate 112-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 17107707-5 2006 On the other hand, repeated administration of corticosterone (for 25 days, the final injection 90 min before contextual fear conditioning training) decreased plasma corticosterone concentration, inhibited exploratory behavior, enhanced freezing responses on retest and produced a complex pattern of changes in c-Fos expression, stimulated by exposure of rats to the aversively conditioned context. Corticosterone 46-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 310-315 17079869-6 2006 The EGCG pretreatment inhibits the Ang II-induced phosphorylation of ERK 1/2, JNK 1/2, or p38 MAPK, and the expression of c-jun or c-fos mRNA. epigallocatechin gallate 4-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-136 17079674-10 2006 DSAP rats displayed significant loss of caudal medullary NA neurons, and markedly blunted Fos activation in the BNST and in corticotropin-releasing hormone-positive PVNmp neurons after YO. dsap 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 17027777-6 2006 Fos-like immunoreactivity (FLI) in rostral, intermediate and caudal portions of dorsomedial (dmPAG), dorsolateral (dlPAG), lateral (lPAG) and ventrolateral (vlPAG) PAG were quantified by a computerized system. dmpag 93-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 16917819-0 2006 Differential c-Fos immunoreactivity in arousal-promoting cell groups following systemic administration of caffeine in rats. Caffeine 106-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 16917819-2 2006 By using c-Fos immunohistochemistry as a marker of neuronal activation, we recently showed that stimulant doses of caffeine activate arousal-promoting hypothalamic orexin (hypocretin) neurons. Caffeine 115-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 16917819-6 2006 The three doses of caffeine induced distinct dose-related patterns of c-Fos immunoreactivity in several arousal-promoting areas, including orexin neurons and adjacent neurons containing neither orexin nor melanin-concentrating hormone; tuberomammillary histaminergic neurons; locus coeruleus noradrenergic neurons; noncholinergic basal forebrain neurons that do not contain parvalbumin; and nondopaminergic neurons in the ventral tegmental area. Caffeine 19-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 16917819-8 2006 Saline controls exhibited only few c-Fos-positive cells in most of the cell groups examined. Sodium Chloride 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 16917819-9 2006 These results indicate that motor-stimulatory doses of caffeine induce a remarkably restricted pattern of c-Fos expression in the arousal-promoting system and suggest that this specific neuronal activation may be involved in the behavioral arousal by caffeine. Caffeine 55-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 16917819-9 2006 These results indicate that motor-stimulatory doses of caffeine induce a remarkably restricted pattern of c-Fos expression in the arousal-promoting system and suggest that this specific neuronal activation may be involved in the behavioral arousal by caffeine. Caffeine 251-259 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 16919249-4 2006 We observed that intensely labeled (type 1) NADPH-d positive neurons were mainly located in the rostral part of the PB; they extended long processes adjacent Fos-IR neurons, but no Fos/type 1 NADPH-d double-labeled neurons were seen. NADP 44-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-161 16919249-6 2006 Additionally, a large number of FG/Fos double-labeled neurons were observed to be surrounded closely by the intensive NADPH-d staining in the el of the PB. [[(2R,3R,4R,5R)-5-(6-aminopurin-9-yl)-3-hydroxy-4-phosphonooxyoxolan-2-yl]methoxy-hydroxyphosphoryl] [(2R,3S,4R,5R)-5-(3-carbamoyl-4-deuterio-4H-pyridin-1-yl)-3,4-dihydroxyoxolan-2-yl]methyl hydrogen phosphate 118-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 16644908-9 2006 Bombesin (8 microg/kg) with ghrelin injected intraperitoneally induced Fos expression in 22.4 +/- 0.8% of CRF-immunoreactive neurons in the PVN. Ghrelin 28-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 16901513-0 2006 Neuroanatomical and pharmacological assessment of Fos expression induced in the rat brain by the phosphodiesterase-4 inhibitor 6-(4-pyridylmethyl)-8-(3-nitrophenyl) quinoline. 6-(4-pyridylmethyl)-8-(3-nitrophenyl)quinoline 127-174 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 16935272-4 2006 After 7 days of treatment with deoxycorticosterone (2 mg/day), an increased number of HSD2 neurons became immunoreactive for the neuronal activity marker c-Fos. Desoxycorticosterone 31-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 16855178-5 2006 In healthy rats, ATB-429 dose dependently (25, 50, or 100 mg/kg) attenuated CRD-induced hypersensitivity and significantly inhibited CRD-induced overexpression of spinal c-FOS mRNA, whereas mesalamine had no effect. 4-anisyltetrazolium blue 17-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-175 16855178-9 2006 Colonic cyclooxygenase-2 and interkeukin-1beta mRNA and spinal c-FOS mRNA expression were significantly down-regulated by ATB-429, but not by mesalamine. 4-anisyltetrazolium blue 122-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 16809001-10 2006 In segments T(13)-L(1), the numbers of Fos-immunoreactive neurons were significantly reduced after treatment with SP-SAP plus baclofen in both dorsal horn regions, and in the deep dorsal horn after baclofen treatment. sp-sap 114-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 16626845-3 2006 Here the authors examine how acute blockage of the NMDA receptor with sub-anaesthetic doses of ketamine affects behavioural assays of fear-conditioned stress (e.g. freezing) and cFos expression in a network of brain areas that have previously been implicated in fear processing. Ketamine 95-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 178-182 16626845-7 2006 Ketamine abolished such increases in cFos expression in most brain areas investigated. Ketamine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-41 16809001-10 2006 In segments T(13)-L(1), the numbers of Fos-immunoreactive neurons were significantly reduced after treatment with SP-SAP plus baclofen in both dorsal horn regions, and in the deep dorsal horn after baclofen treatment. Baclofen 126-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 16809001-10 2006 In segments T(13)-L(1), the numbers of Fos-immunoreactive neurons were significantly reduced after treatment with SP-SAP plus baclofen in both dorsal horn regions, and in the deep dorsal horn after baclofen treatment. Baclofen 198-206 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 16861172-1 2006 The study was designed to determine whether the protein kinase C (PKC) is involved in nociceptive c-Fos expression and the concomitant signaling processes of endogenous opioid-like substances (OLS) that modulate c-Fos expression in the spinal dorsal horn following formalin injection into the unilateral hindpaw in rats by using immunocytochemical techniques. N-biotin-C-Co4(mu3-O)4(Py)4(H2O)4-beta-alanine 193-196 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 212-217 16948600-1 2006 OBJECTIVE: To investigate the effect of adrenalectomy on cholecystokinin-8 (CCK-8)-induced Fos-like immunoreactivity (Fos-LI) in the myenteric neurons of the dorsal vagal complex (DVC) in rats. cholecystokinin 8 76-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-94 16948600-1 2006 OBJECTIVE: To investigate the effect of adrenalectomy on cholecystokinin-8 (CCK-8)-induced Fos-like immunoreactivity (Fos-LI) in the myenteric neurons of the dorsal vagal complex (DVC) in rats. cholecystokinin 8 76-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 16948600-7 2006 RESULTS: After adrenalectomy, CCK-8-induced Fos-LI was attenuated only in the myenteric neurons of the duodenum. cholecystokinin 8 30-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 16861172-3 2006 chelerythrine (Chel), an inhibitor of PKC, the nociceptive c-Fos-like immunoreactive (Fos-LI) neurons in the lumbar dorsal horn ipsilateral to the formalin injection were significantly suppressed with a reduction rate of 60.3% (p < .001) as compared to that in the control group with i.t. chelerythrine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 16861172-3 2006 chelerythrine (Chel), an inhibitor of PKC, the nociceptive c-Fos-like immunoreactive (Fos-LI) neurons in the lumbar dorsal horn ipsilateral to the formalin injection were significantly suppressed with a reduction rate of 60.3% (p < .001) as compared to that in the control group with i.t. chelerythrine 15-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 16861172-11 2006 These results suggest that: (1) PKC may play an important role in the induction of nociceptive c-Fos expression; (2) nociceptive c-Fos expression is subject to the modulation of endogenous OLS that suppress the nociceptive responses of the dorsal horn neurons; and (3) PKC may not be involved in the signaling processes by which the endogenous OLS modulate the nociceptive c-Fos expression in the spinal level. N-biotin-C-Co4(mu3-O)4(Py)4(H2O)4-beta-alanine 189-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 16775195-6 2006 Furthermore, retigabine inhibited dopamine synthesis and c-Fos expression in the striatum under basal conditions. ezogabine 13-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 16861172-11 2006 These results suggest that: (1) PKC may play an important role in the induction of nociceptive c-Fos expression; (2) nociceptive c-Fos expression is subject to the modulation of endogenous OLS that suppress the nociceptive responses of the dorsal horn neurons; and (3) PKC may not be involved in the signaling processes by which the endogenous OLS modulate the nociceptive c-Fos expression in the spinal level. N-biotin-C-Co4(mu3-O)4(Py)4(H2O)4-beta-alanine 189-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 16861172-11 2006 These results suggest that: (1) PKC may play an important role in the induction of nociceptive c-Fos expression; (2) nociceptive c-Fos expression is subject to the modulation of endogenous OLS that suppress the nociceptive responses of the dorsal horn neurons; and (3) PKC may not be involved in the signaling processes by which the endogenous OLS modulate the nociceptive c-Fos expression in the spinal level. N-biotin-C-Co4(mu3-O)4(Py)4(H2O)4-beta-alanine 189-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 16861172-11 2006 These results suggest that: (1) PKC may play an important role in the induction of nociceptive c-Fos expression; (2) nociceptive c-Fos expression is subject to the modulation of endogenous OLS that suppress the nociceptive responses of the dorsal horn neurons; and (3) PKC may not be involved in the signaling processes by which the endogenous OLS modulate the nociceptive c-Fos expression in the spinal level. N-biotin-C-Co4(mu3-O)4(Py)4(H2O)4-beta-alanine 344-347 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 16861172-11 2006 These results suggest that: (1) PKC may play an important role in the induction of nociceptive c-Fos expression; (2) nociceptive c-Fos expression is subject to the modulation of endogenous OLS that suppress the nociceptive responses of the dorsal horn neurons; and (3) PKC may not be involved in the signaling processes by which the endogenous OLS modulate the nociceptive c-Fos expression in the spinal level. N-biotin-C-Co4(mu3-O)4(Py)4(H2O)4-beta-alanine 344-347 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 16944837-4 2006 However, persistent TMJ inflammation significantly increased Fos-LI neurons in the nucleus raphe magnus (NRM) induced by subsequent formalin injection of the masseter muscle and hindpaw (70.2% increase and 53.8% increase, respectively, over the control TMJ-saline-injected rats; P < 0.05). Formaldehyde 132-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 16835009-3 2006 The aim of this study was to determine how the noxious stimulant capsaicin affects intracellular dynamics in the dlPAG evidenced by Fos protein immunoreactivity (index of intracellular activation) and the NADPH-d reactivity. Capsaicin 65-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-135 16835009-5 2006 Compared to vehicle, capsaicin (50mg/kg, subcutaneous) significantly increased NADPH-d reactivity and Fos expression along the dlPAG neuraxis. Capsaicin 21-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 16835009-6 2006 However, less than one percent of the capsaicin-induced Fos activation occurred in NADPH-d-positive cells. Capsaicin 38-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 16835009-6 2006 However, less than one percent of the capsaicin-induced Fos activation occurred in NADPH-d-positive cells. [[(2R,3R,4R,5R)-5-(6-aminopurin-9-yl)-3-hydroxy-4-phosphonooxyoxolan-2-yl]methoxy-hydroxyphosphoryl] [(2R,3S,4R,5R)-5-(3-carbamoyl-4-deuterio-4H-pyridin-1-yl)-3,4-dihydroxyoxolan-2-yl]methyl hydrogen phosphate 83-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 16930414-0 2006 Cocaine-induced locomotor activity and Fos expression in nucleus accumbens are sensitized for 6 months after repeated cocaine administration outside the home cage. Cocaine 118-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 16930414-1 2006 Induction of the immediate early gene protein product Fos has been used extensively to assess neural activation in the striatum after repeated cocaine administration to rats in their home cages but rarely after repeated administration outside the home cage, which produces more robust locomotor sensitization. Cocaine 143-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 16930414-2 2006 In the present study, we found cocaine-induced Fos expression in nucleus accumbens, but not caudate-putamen, was enhanced 1 and 6 months after repeated drug administration in locomotor activity chambers. Cocaine 31-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 16930414-4 2006 In contrast, cocaine-induced Fos expression was absent altogether in nucleus accumbens and unaltered in caudate-putamen 1 month after repeated cocaine administration in the home cage. Cocaine 13-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 16930414-6 2006 Bilateral infusions of the GABA agonists baclofen and muscimol (1 microg/side) into nucleus accumbens of sensitized rats blocked cocaine-induced Fos expression and locomotor activity. gamma-Aminobutyric Acid 27-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-148 16777145-5 2006 In control experiments, CHX (3 mg/kg) blocked c-Fos protein expression after foot-shock stress and impaired the acquisition of conditioned freezing but did not inhibit spontaneous locomotor activity and sucrose drinking. Cycloheximide 24-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 16797618-0 2006 Atropine methyl nitrate increases myenteric but not dorsal vagal complex Fos-like immunoreactivity in the rat. methylatropine 0-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-76 16797618-1 2006 Atropine methyl nitrate (AMN, 0.05, 0.5 and 25 mg/kg) intraperitoneally increased Fos-like immunoreactivity (Fos-LI) in the myenteric plexus, but not the dorsal vagal complex (DVC, the area postrema (AP), nucleus of the solitary tract (NTS) and the dorsal motor nucleus of the vagus (DMV)) in adult, male Sprague-Dawley rats. methylatropine 0-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 16797618-1 2006 Atropine methyl nitrate (AMN, 0.05, 0.5 and 25 mg/kg) intraperitoneally increased Fos-like immunoreactivity (Fos-LI) in the myenteric plexus, but not the dorsal vagal complex (DVC, the area postrema (AP), nucleus of the solitary tract (NTS) and the dorsal motor nucleus of the vagus (DMV)) in adult, male Sprague-Dawley rats. methylatropine 0-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-112 16781683-4 2006 Intracerebroventricular administration of beta-guanidinopropionic acid, a compound known to decrease intracellular creatine concentration by competition for uptake, resulted in decreased food intake and body weight and increased Fos expression in the hypothalamus. guanidinopropionic acid 42-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 229-232 16781683-4 2006 Intracerebroventricular administration of beta-guanidinopropionic acid, a compound known to decrease intracellular creatine concentration by competition for uptake, resulted in decreased food intake and body weight and increased Fos expression in the hypothalamus. Creatine 115-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 229-232 16611858-8 2006 FLU had a sharp down-regulation of c-fos that was comparable with all the compounds except CPZ and CLO. Flutamide 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 16516888-1 2006 We investigated the effects of a selective lesion of the substantia nigra pars reticulata (SNr), obtained by stereotaxic injection of ibotenic acid, on the cortical expression of Fos protein induced by striatal infusion of dopamine, D1-like agonist SKF 38393, in Sprague-Dawley rats. Ibotenic Acid 134-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-182 16516888-1 2006 We investigated the effects of a selective lesion of the substantia nigra pars reticulata (SNr), obtained by stereotaxic injection of ibotenic acid, on the cortical expression of Fos protein induced by striatal infusion of dopamine, D1-like agonist SKF 38393, in Sprague-Dawley rats. Dopamine 223-231 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-182 16516888-1 2006 We investigated the effects of a selective lesion of the substantia nigra pars reticulata (SNr), obtained by stereotaxic injection of ibotenic acid, on the cortical expression of Fos protein induced by striatal infusion of dopamine, D1-like agonist SKF 38393, in Sprague-Dawley rats. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 249-258 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-182 16516888-3 2006 The striatal, unilateral infusion of 30 mM SKF 38393 induced consistent Fos expression throughout the whole ipsilateral cerebral cortex, including motor, sensorimotor, associative, and limbic areas; such expression was dramatically reduced by excitotoxic lesion of the ipsilateral SNr. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 43-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 16996246-3 2006 In the current study, the effects of vaccination on nicotine-induced changes in fetal (3)H-epibatidine binding and c-fos mRNA expression were evaluated using tissue from a previous pharmacokinetic study of vaccination. Nicotine 52-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 16996246-6 2006 Gestational nicotine exposure produced significant increases in (125)I-epibatidine binding to brain and spinal cord on GD20, and decreased c-fos mRNA expression in fetal striatum, adrenal and lung. Nicotine 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 16996246-8 2006 These data suggest that nicotine dosing, using a clinically relevant intermittent bolus dose regimen, produces substantial changes in fetal nicotinic receptor and c-fos mRNA expression. Nicotine 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-168 16996246-9 2006 The decrease in c-fos mRNA expression contrasts with previously reported increases, and suggests that the nicotine dosing regimen used may influence its effects. Nicotine 106-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 17063538-0 2006 [Nitric oxide regulates c-fos expression in osteoblastic cells induced by wall-shear]. Nitric Oxide 1-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 17063538-1 2006 OBJECTIVE: To observe regulation of nitric oxide on c-fos expression in osteoblastic cells in response to changes in wall-shear stress in vitro. Nitric Oxide 36-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 17063538-5 2006 The expression of c-fos mRNA was decreased after pre-application with L-NMMA and increased after use of SNP. omega-N-Methylarginine 70-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 17147208-3 2006 Exposure to propionic acid produced a moderate activation of c-Fos expression, mainly in the granular layer of the dorsomedial part of the bulb. propionic acid 12-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 16854390-6 2006 c-Fos expression, determined in the arcuate nucleus of the hypothalamus in the same animals 1 week after the last formalin test, was higher in GDX than INT animals; moreover, while in INT rats, c-Fos was higher in the formalin-injected animals (SF and FF) than in HC, in GDX, it did not differ among groups. Formaldehyde 114-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 16854390-6 2006 c-Fos expression, determined in the arcuate nucleus of the hypothalamus in the same animals 1 week after the last formalin test, was higher in GDX than INT animals; moreover, while in INT rats, c-Fos was higher in the formalin-injected animals (SF and FF) than in HC, in GDX, it did not differ among groups. GDP-alpha-D-mannuronic acid 143-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 16854390-6 2006 c-Fos expression, determined in the arcuate nucleus of the hypothalamus in the same animals 1 week after the last formalin test, was higher in GDX than INT animals; moreover, while in INT rats, c-Fos was higher in the formalin-injected animals (SF and FF) than in HC, in GDX, it did not differ among groups. Formaldehyde 218-226 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 16854390-6 2006 c-Fos expression, determined in the arcuate nucleus of the hypothalamus in the same animals 1 week after the last formalin test, was higher in GDX than INT animals; moreover, while in INT rats, c-Fos was higher in the formalin-injected animals (SF and FF) than in HC, in GDX, it did not differ among groups. GDP-alpha-D-mannuronic acid 271-274 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 16951565-5 2006 Fos expression induced by the alphabeta-methylene ATP injection in dorsal horn neurons was also increased after the pre-application of PAR2 agonists. alpha,beta-methyleneadenosine 5'-triphosphate 30-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 16730913-2 2006 Thus, physiological studies have shown that peripheral immune stimuli, as well as the administration of aversive substances such as lithium chloride, evoke a prominent Fos-expression in the lateral parabrachial nucleus and behavioral experiments have demonstrated that this structure is critical for the acquisition of conditioned taste aversion. Lithium Chloride 132-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-171 16730913-4 2006 Dual-labeling in situ hybridization showed melanocortin-4 receptor expression on neurons in the external lateral parabrachial subnucleus that displayed lipopolysaccharide- or lithium chloride-induced expression of c-fos mRNA. Lithium Chloride 175-191 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 214-219 16844117-6 2006 Icv injections of CBD (10 microg/5microl) induced an enhancement of c-Fos expression in waking-related brain areas such as hypothalamus and dorsal raphe nucleus (DRD). Cannabidiol 18-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 16930414-6 2006 Bilateral infusions of the GABA agonists baclofen and muscimol (1 microg/side) into nucleus accumbens of sensitized rats blocked cocaine-induced Fos expression and locomotor activity. Baclofen 41-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-148 16930414-6 2006 Bilateral infusions of the GABA agonists baclofen and muscimol (1 microg/side) into nucleus accumbens of sensitized rats blocked cocaine-induced Fos expression and locomotor activity. Muscimol 54-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-148 16930414-6 2006 Bilateral infusions of the GABA agonists baclofen and muscimol (1 microg/side) into nucleus accumbens of sensitized rats blocked cocaine-induced Fos expression and locomotor activity. Cocaine 129-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-148 16890608-8 2006 Indomethacin-induced acute ileitis led to repression of ASBT in wild-type mice and in the transgenic rat ASBT promoter reporter, while paradoxical activation of ASBT was seen in c-fos-null mice. Indomethacin 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 178-183 16739166-5 2006 Film autoradiographic studies showed that nicotine significantly increased c-fos mRNA expression in both PVN and CEA. Nicotine 42-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 16739166-6 2006 Pretreatment with the centrally acting nicotinic antagonist, mecamylamine (1 mg/kg), blocked nicotine"s effects, whereas pretreatment with the peripherally acting antagonist, hexamethonium (5 mg/kg), did not, indicating that c-fos induction was mediated by a central nicotinic receptor. Nicotine 93-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 225-230 16739166-7 2006 Double labeling studies showed that nicotine induced c-fos expression within CRF cells in the PVN, as well as in a small population of ENK cells, but not in PVN DYN cells. Nicotine 36-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 16739166-8 2006 In contrast, there was no significant nicotine-induced increase in c-fos expression in CEA CRF or DYN cells, whereas nicotine treatment did increase c-fos expression within CEA ENK cells. Nicotine 117-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 149-154 16690725-8 2006 LY466195 was also efficacious in the c-fos migraine model, with a dose of 1 microg/kg i.v. 6-((2-carboxy-4,4-difluoro-1-pyrrolidinyl)methyl)decahydro-3-isoquinolinecarboxylic acid 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 16690725-11 2006 The diethyl ester prodrug of LY466195 was also efficacious in the same PPE and c-fos models after oral administration at doses of 10 and 100 microg/kg, respectively while having no N-methyl-D-aspartate antagonist-like behavioral effects at oral doses up to 100 mg/kg. Ether 4-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 16690725-11 2006 The diethyl ester prodrug of LY466195 was also efficacious in the same PPE and c-fos models after oral administration at doses of 10 and 100 microg/kg, respectively while having no N-methyl-D-aspartate antagonist-like behavioral effects at oral doses up to 100 mg/kg. 6-((2-carboxy-4,4-difluoro-1-pyrrolidinyl)methyl)decahydro-3-isoquinolinecarboxylic acid 29-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 16712881-8 2006 U0126 in the spinal cord were accompanied by decreased scores of morphine withdrawal and the inhibited spinal Fos protein (a maker for neuronal excitation or activation) expression induced by morphine withdrawal. U 0126 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 16712881-8 2006 U0126 in the spinal cord were accompanied by decreased scores of morphine withdrawal and the inhibited spinal Fos protein (a maker for neuronal excitation or activation) expression induced by morphine withdrawal. Morphine 192-200 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 16730124-5 2006 Noxious stimulation of the occipital muscle in rat using mustard oil and mineral oil produced significantly altered Fos expression in the trigeminocervical complex compared with the surgical control (H(4)=31.3, P<0.001, Kruskal-Wallis). mustard oil 57-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 16730124-5 2006 Noxious stimulation of the occipital muscle in rat using mustard oil and mineral oil produced significantly altered Fos expression in the trigeminocervical complex compared with the surgical control (H(4)=31.3, P<0.001, Kruskal-Wallis). Mineral Oil 73-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 16730124-6 2006 Baseline expression was 11 (median, range 4, 17) fos positive cells in the trigeminocervical complex, occipital muscle treated with mustard oil produced 23 (17, 33) and mineral oil a smaller effect of 19 (15, 25) fos positive cells, respectively (P=0.046). mustard oil 132-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 16730124-6 2006 Baseline expression was 11 (median, range 4, 17) fos positive cells in the trigeminocervical complex, occipital muscle treated with mustard oil produced 23 (17, 33) and mineral oil a smaller effect of 19 (15, 25) fos positive cells, respectively (P=0.046). mustard oil 132-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-216 16730124-6 2006 Baseline expression was 11 (median, range 4, 17) fos positive cells in the trigeminocervical complex, occipital muscle treated with mustard oil produced 23 (17, 33) and mineral oil a smaller effect of 19 (15, 25) fos positive cells, respectively (P=0.046). Mineral Oil 169-180 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 16867172-3 2006 The aim of the present study was to use c-fos gene expression as a measure of nociceptive input after intramuscular injection of different oxytetracycline formulations. Oxytetracycline 139-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 16867172-11 2006 Rats receiving an injection of Engemycin had 1,932+/-893 Fos-positive neurones, which was not significantly different from the saline group. Oxytetracycline 31-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 16565164-30 2006 or i.c.v., buprenorphine decreased the number of Fos-like immunoreactivity positive neurons in the L4-L5 spinal dorsal horn. Buprenorphine 11-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 16631212-0 2006 Effects of chronic nicotine administration and its withdrawal on striatal FosB/DeltaFosB and c-Fos expression in rats and mice. Nicotine 19-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 16631212-4 2006 In rats given nicotine subcutaneously once daily for 5days FosB/DeltaFosB expression was elevated in the NAcc still after 24-h withdrawal suggesting accumulation of DeltaFosB but in the CPu neither FosB/DeltaFosB nor c-Fos expression was altered. Nicotine 14-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 217-222 16678862-0 2006 Fos immunolabelling evidence for brain regions involved in the Pavlovian degraded contingency effect and in its disruption by atropine. Atropine 126-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 16678862-5 2006 Compared to contingent conditioning, the DCE was associated with a decrease of the amount of Fos immunoreactive neurons within the auditory system and the amygdala and an increase within the medial prefrontal cortex (mPFC). ethylene dichloride 41-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 16678862-6 2006 Compared to the normal DCE, atropine-induced disruption of the DCE was associated with an increase of the amount of Fos immunoreactive neurons within the central nucleus of the amygdala. Atropine 28-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 16678862-6 2006 Compared to the normal DCE, atropine-induced disruption of the DCE was associated with an increase of the amount of Fos immunoreactive neurons within the central nucleus of the amygdala. ethylene dichloride 63-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 16678862-7 2006 When atropine-induced suppression of the DCE, Fos pattern was modified in the mPFC with a change in Fos immunoreactivity, but no longer associated with the DCE. Atropine 5-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 16678862-7 2006 When atropine-induced suppression of the DCE, Fos pattern was modified in the mPFC with a change in Fos immunoreactivity, but no longer associated with the DCE. Atropine 5-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-103 16678862-7 2006 When atropine-induced suppression of the DCE, Fos pattern was modified in the mPFC with a change in Fos immunoreactivity, but no longer associated with the DCE. ethylene dichloride 41-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 16678862-7 2006 When atropine-induced suppression of the DCE, Fos pattern was modified in the mPFC with a change in Fos immunoreactivity, but no longer associated with the DCE. ethylene dichloride 41-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-103 16837891-3 2006 In this study, we examined the effects of fructo-oligosaccharides (FOS), a prebiotic, on enhancing the effects of soy isoflavones on bone in ovariectomized osteopenic female rats. fructooligosaccharide 42-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 16837891-3 2006 In this study, we examined the effects of fructo-oligosaccharides (FOS), a prebiotic, on enhancing the effects of soy isoflavones on bone in ovariectomized osteopenic female rats. Isoflavones 118-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 16580740-4 2006 Several reports show cocaine-induced c-fos expression particularly in the intermediate zone after 14, but not 2, drug-free days following repeated cocaine administration, suggesting that this region may be involved in sensitization and particularly in the later phase of expression, versus the earlier phase of sensitization. Cocaine 21-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 16580740-4 2006 Several reports show cocaine-induced c-fos expression particularly in the intermediate zone after 14, but not 2, drug-free days following repeated cocaine administration, suggesting that this region may be involved in sensitization and particularly in the later phase of expression, versus the earlier phase of sensitization. Cocaine 147-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 16807338-5 2006 Using double immunocytochemistry, we found that rats exhibiting panic-like responses (e.g., L-AG plus lactate) had increased c-Fos immunoreactivity in DMH neurons expressing the NMDA receptor 1 (NR1) subunit, but not those expressing the glutamate receptor 2 and 3 subunits of the AMPA receptors. l-ag 92-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 16807338-5 2006 Using double immunocytochemistry, we found that rats exhibiting panic-like responses (e.g., L-AG plus lactate) had increased c-Fos immunoreactivity in DMH neurons expressing the NMDA receptor 1 (NR1) subunit, but not those expressing the glutamate receptor 2 and 3 subunits of the AMPA receptors. Lactic Acid 102-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 16807338-5 2006 Using double immunocytochemistry, we found that rats exhibiting panic-like responses (e.g., L-AG plus lactate) had increased c-Fos immunoreactivity in DMH neurons expressing the NMDA receptor 1 (NR1) subunit, but not those expressing the glutamate receptor 2 and 3 subunits of the AMPA receptors. 1,2-Dimethylhydrazine 151-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 16716266-5 2006 To determine whether opioid receptor ligands administered into the rLHa affect neuronal activation in this brain site, we studied cFos immunoreactivity (cFos IR) in response to rLHa stimulation with naltrexone. Naltrexone 199-209 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-134 16716266-5 2006 To determine whether opioid receptor ligands administered into the rLHa affect neuronal activation in this brain site, we studied cFos immunoreactivity (cFos IR) in response to rLHa stimulation with naltrexone. Naltrexone 199-209 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-157 16725125-0 2006 Repeated l-DOPA treatment increases c-fos and BDNF mRNAs in the subthalamic nucleus in the 6-OHDA rat model of Parkinson"s disease. Levodopa 9-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 16725125-0 2006 Repeated l-DOPA treatment increases c-fos and BDNF mRNAs in the subthalamic nucleus in the 6-OHDA rat model of Parkinson"s disease. Oxidopamine 91-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 16725125-2 2006 This study used the unilateral 6-OHDA rat model of Parkinson"s disease to examine effects of l-DOPA on the expression of c-fos and BDNF mRNAs in these nuclei. Levodopa 93-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-126 16725125-4 2006 Both a single and repeated injections of l-DOPA induced c-fos, but not BDNF, in the dopamine-depleted striatum. Levodopa 41-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 16725125-4 2006 Both a single and repeated injections of l-DOPA induced c-fos, but not BDNF, in the dopamine-depleted striatum. Dopamine 84-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 16725125-5 2006 However, repeated l-DOPA treatment increased c-fos and BDNF in the dopamine-depleted subthalamic nucleus. Levodopa 18-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 16725125-5 2006 However, repeated l-DOPA treatment increased c-fos and BDNF in the dopamine-depleted subthalamic nucleus. Dopamine 67-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 16615128-7 2006 Systemic administration of morphine significantly suppressed CFA-induced Fos in the PAG in males only. Morphine 27-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-76 16696958-5 2006 Immunoblot and immunohistochemical analysis demonstrated that ERK-c-Fos signal transduction pathway was activated by exogenous 17beta-estradiol in ganglion cell layer. Estradiol 127-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 16569443-6 2006 The results obtained showed that freezing behavior induced by semicarbazide was associated with an increase in Fos expression in the dorsomedial column of the PAG (dmPAG) only, while bicuculline-induced escape was related to widespread increase in Fos labeling, notably in the periaqueductal gray, hypothalamus nuclei, amygdaloid nuclei, the laterodorsal nucleus of thalamus (LD), the cuneiform nucleus (CnF) and the locus coeruleus (LC). carbamylhydrazine 62-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 16569443-6 2006 The results obtained showed that freezing behavior induced by semicarbazide was associated with an increase in Fos expression in the dorsomedial column of the PAG (dmPAG) only, while bicuculline-induced escape was related to widespread increase in Fos labeling, notably in the periaqueductal gray, hypothalamus nuclei, amygdaloid nuclei, the laterodorsal nucleus of thalamus (LD), the cuneiform nucleus (CnF) and the locus coeruleus (LC). phenylacetylglycine 159-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 16569443-6 2006 The results obtained showed that freezing behavior induced by semicarbazide was associated with an increase in Fos expression in the dorsomedial column of the PAG (dmPAG) only, while bicuculline-induced escape was related to widespread increase in Fos labeling, notably in the periaqueductal gray, hypothalamus nuclei, amygdaloid nuclei, the laterodorsal nucleus of thalamus (LD), the cuneiform nucleus (CnF) and the locus coeruleus (LC). dmpag 164-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 16569443-6 2006 The results obtained showed that freezing behavior induced by semicarbazide was associated with an increase in Fos expression in the dorsomedial column of the PAG (dmPAG) only, while bicuculline-induced escape was related to widespread increase in Fos labeling, notably in the periaqueductal gray, hypothalamus nuclei, amygdaloid nuclei, the laterodorsal nucleus of thalamus (LD), the cuneiform nucleus (CnF) and the locus coeruleus (LC). Bicuculline 183-194 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 248-251 16732098-7 2006 Finally, epidural morphine (30 microg/10 microl) in oxytocin-treated rats, although resulting in no change of labor duration, significantly decreased the number of stretches (8 +/- 2 vs. 57 +/- 12 for epidural saline) and the number of c-Fos-positive neurons in the lumbosacral spinal segments (80 +/- 25 vs. 165 +/- 17 for epidural saline). Morphine 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 236-241 16944776-10 2006 Sodium selenite at the doses of 5, 10, and 20 micromol/kg caused c-myc, c-fos, and c-jun overexpression obviously. Sodium Selenite 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 16944776-15 2006 CONCLUSION: Selenium at 5-20 micromol/kg can induce DNA damage, apoptosis, and overexpression of c-myc, c-fos, and c-jun in rat hepatocytes. Selenium 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 16543367-6 2006 Coexpression of the cAMP-responsive element-binding protein-binding protein and steroid receptor coactivator-1a further enhanced the Fos/Jun-mediated transcription. Cyclic AMP 20-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-136 15994217-9 2006 RESULTS: Treatment with NOX-B11 30 nmol suppressed ghrelin induced c-Fos-like immunoreactivity in the arcuate nucleus and blocked the ghrelin induced increase in food intake within the first half hour after ghrelin injection (mean 1.13 (SEM 0.59) g/kg body weight; 4.94 (0.63) g/kg body weight versus 0.58 (0.58) g/kg body weight; p<0.0001). NOX-B11 24-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 16547969-7 2006 C-fos antisense study also revealed that depression of Fos-PPE signaling in the ipsi-MVN caused significantly more severe behavioral deficits during vestibular compensation. ipsi-mvn 80-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 16549369-0 2006 Functional magnetic resonance imaging and c-Fos mapping in rats following an anorectic dose of m-chlorophenylpiperazine. 1-(3-chlorophenyl)piperazine 95-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 16549369-2 2006 An anorectic dose of the 5-HT(1B/2C) receptor agonist m-chlorophenylpiperazine (mCPP; 3 mg/kg s.c.) was used to compare BOLD contrast fMRI with expression of the c-Fos protein. 1-(3-chlorophenyl)piperazine 54-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 16547969-7 2006 C-fos antisense study also revealed that depression of Fos-PPE signaling in the ipsi-MVN caused significantly more severe behavioral deficits during vestibular compensation. ipsi-mvn 80-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 16504219-0 2006 L-3,4-dihydroxyphenylalanine-induced c-Fos expression in the CNS under inhibition of central aromatic L-amino acid decarboxylase. Levodopa 0-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 16504219-2 2006 To map the DOPAergic system functionally, DOPA-induced c-Fos expression was detected under inhibition of central aromatic L-amino acid decarboxylase (AADC). Levodopa 11-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 16678866-7 2006 Further, our results suggest that the enhanced behavioural changes after AMPH and PCP administration were associated with increased expression of AP-1 proteins (Fos and Jun) in the cortex, striatum and hippocampus and that their binding to AP-1 sites on the DNA contributes to long-term changes in rat brain. Amphetamine 73-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-164 16504219-3 2006 In rats treated with a central AADC inhibitor, DOPA significantly increased the number of c-Fos-positive nuclei in the paraventricular nuclei (PVN) and the nucleus tractus solitarii (NTS), and showed a tendency to increase in the supraoptic nuclei (SON), but not in the striatum. Levodopa 47-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 16678866-7 2006 Further, our results suggest that the enhanced behavioural changes after AMPH and PCP administration were associated with increased expression of AP-1 proteins (Fos and Jun) in the cortex, striatum and hippocampus and that their binding to AP-1 sites on the DNA contributes to long-term changes in rat brain. Phencyclidine 82-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-164 16504219-4 2006 On the other hand, DOPA with a peripheral AADC inhibitor elevated the level of c-Fos-positive nuclei in the four regions, suggesting that DOPA itself induces c-Fos expression in the SON, PVN and NTS. Levodopa 19-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 16504219-4 2006 On the other hand, DOPA with a peripheral AADC inhibitor elevated the level of c-Fos-positive nuclei in the four regions, suggesting that DOPA itself induces c-Fos expression in the SON, PVN and NTS. Levodopa 19-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 16504219-4 2006 On the other hand, DOPA with a peripheral AADC inhibitor elevated the level of c-Fos-positive nuclei in the four regions, suggesting that DOPA itself induces c-Fos expression in the SON, PVN and NTS. Levodopa 138-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 16504219-4 2006 On the other hand, DOPA with a peripheral AADC inhibitor elevated the level of c-Fos-positive nuclei in the four regions, suggesting that DOPA itself induces c-Fos expression in the SON, PVN and NTS. Levodopa 138-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 16504219-5 2006 In rats treated with 6-hydroxydopamine (6-OHDA) to lesion the nigrostriatal dopamine (DA) pathway, DOPA significantly induced c-Fos expression in the four regions under the inhibition of peripheral AADC. Oxidopamine 21-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 16504219-5 2006 In rats treated with 6-hydroxydopamine (6-OHDA) to lesion the nigrostriatal dopamine (DA) pathway, DOPA significantly induced c-Fos expression in the four regions under the inhibition of peripheral AADC. Oxidopamine 40-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 16504219-5 2006 In rats treated with 6-hydroxydopamine (6-OHDA) to lesion the nigrostriatal dopamine (DA) pathway, DOPA significantly induced c-Fos expression in the four regions under the inhibition of peripheral AADC. Dopamine 30-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 16504219-5 2006 In rats treated with 6-hydroxydopamine (6-OHDA) to lesion the nigrostriatal dopamine (DA) pathway, DOPA significantly induced c-Fos expression in the four regions under the inhibition of peripheral AADC. Dopamine 44-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 16504219-5 2006 In rats treated with 6-hydroxydopamine (6-OHDA) to lesion the nigrostriatal dopamine (DA) pathway, DOPA significantly induced c-Fos expression in the four regions under the inhibition of peripheral AADC. Levodopa 99-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 16616405-10 2006 Nevertheless, exposure to B(a)P plus inhaled SO2 increased the mRNA and protein levels of c-jun and c-fos in lungs compared with lungs exposed to SO2 alone. Sulfur Dioxide 45-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 16859115-10 2006 The expressions of epithelial pERK and c-fos in the NGF group were significantly higher than those in the control group, and PD98059 could inhibit NGF inducing NHBEC to produce pERK and c-fos. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 125-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 186-191 16927803-7 2006 The expression of c-fos and NR2A immunoreactive neurons in auditory cortexes of the sodium salicylate group was significantly higher than that of other two control groups. Sodium Salicylate 84-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 16543266-0 2006 Oestrogen and weight loss decrease isoproterenol-induced Fos immunoreactivity and angiotensin type 1 mRNA in the subfornical organ of female rats. Isoproterenol 35-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 16543266-4 2006 The goal of this study was to evaluate the contributions of oestrogen and weight loss to isoproterenol (isoprenaline; Iso)-induced Fos immunoreactivity (IR) and to angiotensin type 1 (AT1) receptor mRNA in forebrain regions implicated in the control of fluid balance. Isoproterenol 89-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-134 16543266-4 2006 The goal of this study was to evaluate the contributions of oestrogen and weight loss to isoproterenol (isoprenaline; Iso)-induced Fos immunoreactivity (IR) and to angiotensin type 1 (AT1) receptor mRNA in forebrain regions implicated in the control of fluid balance. Isoproterenol 104-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-134 16543266-4 2006 The goal of this study was to evaluate the contributions of oestrogen and weight loss to isoproterenol (isoprenaline; Iso)-induced Fos immunoreactivity (IR) and to angiotensin type 1 (AT1) receptor mRNA in forebrain regions implicated in the control of fluid balance. Isoproterenol 118-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-134 16543266-5 2006 Isoproterenol significantly increased Fos IR in the hypothalamic paraventricular and supraoptic nuclei, the subfornical organ (SFO), and the organum vasculosum of the lamina terminalis, but had no effect on AT1 mRNA expression. Isoproterenol 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 16600401-1 2006 Chemical sympathectomy with daily, intraperitoneal (IP) injections of guanethidine sulfate to adult rats, attenuated myenteric, but not dorsal vagal complex (DVC) Fos-like immunoreactivity (Fos-LI) by cholecystokinin-8 (CCK). Guanethidine 70-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 16600401-1 2006 Chemical sympathectomy with daily, intraperitoneal (IP) injections of guanethidine sulfate to adult rats, attenuated myenteric, but not dorsal vagal complex (DVC) Fos-like immunoreactivity (Fos-LI) by cholecystokinin-8 (CCK). Guanethidine 70-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 190-193 16446041-11 2006 By examining expression of Fos protein in response to intraplantar injection of formaldehyde we provide evidence that many of the preprotachykinin B cells in lamina I and the outer part of lamina II respond to noxious stimulation. Formaldehyde 80-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 16630589-4 2006 To test this hypothesis, we counted Fos-LI in the DVC of ondansetron (1 mg/kg; 5-HT3 receptor antagonist) and vehicle-treated rats following gastric balloon distension (5 ml), CCK (1 microg/kg) administration, or CCK combined with gastric distension. Ondansetron 57-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-39 16630589-5 2006 Ondansetron administration attenuated DVC Fos-LI by CCK administration. Ondansetron 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 16630589-6 2006 Likewise, ondansetron attenuated Fos-LI by gastric distension in the DVC, specifically within the nucleus of the solitary tract (NTS) and area postrema (AP) nuclei. Ondansetron 10-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 16630589-7 2006 The most pronounced attenuation of distension-induced Fos-LI by ondansetron occurred in the NTS, particularly in the medial and intermedial NTS. Ondansetron 64-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 16630589-9 2006 Furthermore, ondansetron administration attenuated the overall DVC enhanced Fos-LI induced by CCK + gastric distension, in particular at the NTS and AP nuclei. Ondansetron 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 16630589-9 2006 Furthermore, ondansetron administration attenuated the overall DVC enhanced Fos-LI induced by CCK + gastric distension, in particular at the NTS and AP nuclei. cyqualon 63-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 16306162-0 2006 Differential effects of water and saline intake on water deprivation-induced c-Fos staining in the rat. Water 24-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 16306162-0 2006 Differential effects of water and saline intake on water deprivation-induced c-Fos staining in the rat. Sodium Chloride 34-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 16306162-0 2006 Differential effects of water and saline intake on water deprivation-induced c-Fos staining in the rat. Water 51-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 16306162-6 2006 Rehydration with water significantly decreased AVP levels and Fos staining in the SON, PVN, and RVL and significantly increased Fos expression in the perinuclear zone of the SON, NTS, and parabrachial nucleus. Water 17-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 16306162-6 2006 Rehydration with water significantly decreased AVP levels and Fos staining in the SON, PVN, and RVL and significantly increased Fos expression in the perinuclear zone of the SON, NTS, and parabrachial nucleus. Water 17-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-131 16306162-11 2006 Rehydration with water or saline produces differential effects on plasma AVP, Fos staining, and sodium concentration. Water 17-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 16306162-11 2006 Rehydration with water or saline produces differential effects on plasma AVP, Fos staining, and sodium concentration. Sodium Chloride 26-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 16399878-6 2006 Induction of necrotizing pancreatitis by treatment with L-arginine caused a 12-fold increase in the number of spinal neurons expressing the proto-oncogene c-fos in laminae I and II of L1, suggesting activation of nociceptive pathways. Arginine 56-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-160 16399878-8 2006 Systemic administration of the TRPV1 antagonist capsazepine inhibited c-fos expression by 2.5-fold and abdominal contractions by 4-fold. capsazepine 48-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 16616732-4 2006 Furthermore, capsaicin activated more Fos-positive cells than vehicle within all subregions of the DRN but with a caudal versus rostral predominance in activation pattern. Capsaicin 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 16616732-5 2006 In addition, a high proportion of capsaicin-induced Fos cells in the midline but almost none in lateral wing stained for NADPH-d. Capsaicin 34-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 16626641-2 2006 The role of capsaicin-sensitive afferents in Fos and gastric responses to distension was also investigated. Capsaicin 12-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 16626641-6 2006 Pretreatment with systemic capsaicin prevented both the brain increase in Fos expression and the inhibition of gastric emptying induced by the colon distension. Capsaicin 27-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 16730785-11 2006 Furthermore, pretreatment with chelerythrine significantly inhibited the expressions of c-fos-LI evoked by alpha-m-5-HT in laminae I-VI and by 5-HT in laminae I-II. chelerythrine 31-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 16706847-0 2006 L-DOPA-induced dyskinesia in adult rats with a unilateral 6-OHDA lesion of dopamine neurons is paralleled by increased c-fos gene expression in the subthalamic nucleus. Levodopa 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 16706847-6 2006 The objective of this work was to study the effects of acute or chronic systemic administration of L-DOPA to adult rats with a unilateral 6-hydroxydopamine (6-OHDA) lesion of dopamine neurons on c-fos expression in the STN and test the hypothesis that these effects correlate with L-DOPA-induced dyskinesias. Levodopa 99-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-200 16706847-6 2006 The objective of this work was to study the effects of acute or chronic systemic administration of L-DOPA to adult rats with a unilateral 6-hydroxydopamine (6-OHDA) lesion of dopamine neurons on c-fos expression in the STN and test the hypothesis that these effects correlate with L-DOPA-induced dyskinesias. Oxidopamine 138-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-200 16706847-6 2006 The objective of this work was to study the effects of acute or chronic systemic administration of L-DOPA to adult rats with a unilateral 6-hydroxydopamine (6-OHDA) lesion of dopamine neurons on c-fos expression in the STN and test the hypothesis that these effects correlate with L-DOPA-induced dyskinesias. Oxidopamine 157-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-200 16706847-6 2006 The objective of this work was to study the effects of acute or chronic systemic administration of L-DOPA to adult rats with a unilateral 6-hydroxydopamine (6-OHDA) lesion of dopamine neurons on c-fos expression in the STN and test the hypothesis that these effects correlate with L-DOPA-induced dyskinesias. Dopamine 147-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-200 16706847-8 2006 Our results confirm earlier evidence that the chronic administration of L-DOPA to rats with a unilateral 6-OHDA lesion increases c-fos expression in the STN. Levodopa 72-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 16706847-8 2006 Our results confirm earlier evidence that the chronic administration of L-DOPA to rats with a unilateral 6-OHDA lesion increases c-fos expression in the STN. Oxidopamine 105-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 16706847-9 2006 We also report that c-fos expression can be increased following an acute injection of L-DOPA to 6-OHDA-lesioned rats but not following a chronic injection of L-DOPA to sham-operated, unlesioned rats. Levodopa 86-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 16706847-9 2006 We also report that c-fos expression can be increased following an acute injection of L-DOPA to 6-OHDA-lesioned rats but not following a chronic injection of L-DOPA to sham-operated, unlesioned rats. Oxidopamine 96-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 16498678-7 2006 In addition, CTb/Fos double-labeled cells constituted 43% of all the singly CTb-labeled cells counted in the DPGi compared with 29% for the LPGi, 18% for the rostral PAG, and 10% or less for the other structures. lpgi 140-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 16448750-0 2006 GSM radiation triggers seizures and increases cerebral c-Fos positivity in rats pretreated with subconvulsive doses of picrotoxin. Picrotoxin 119-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 16448750-3 2006 Non-irradiated picrotoxin-treated rats did not suffer seizures, and their cerebral c-Fos counts were significantly lower. Picrotoxin 15-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 16413065-5 2006 Microinjection of l-glutamate (2-5 nmol) into the PVN produced a frequent yawning accompanied by an arousal shift in the ECoG, and these behavioral effects were associated with a significant increase of c-Fos positive CRF neurons in the medial parvocellular subdivision of the PVN. Glutamic Acid 18-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 203-208 16406667-0 2006 5-HT1A receptor activation counteracts c-Fos immunoreactivity induced in serotonin neurons of the raphe nuclei after immobilization stress in the male rat. Serotonin 73-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 17704842-0 2006 Effect of water restrictions on the physiological parameters, psychological behavior and brain c-Fos expression in rats. Water 10-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 17704842-11 2006 The changes of Fos expression in most of nuclei in EB group began at day 3, at least persisted till day 7, and backed down at day 14, while in WR group, similar changes started at day 7 and reached its peak at day 14. Eriochrome Blue SE 51-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-18 16084549-0 2006 Activations of c-fos/c-jun signaling are involved in the modulation of hypothalamic superoxide dismutase (SOD) and neuropeptide Y (NPY) gene expression in amphetamine-mediated appetite suppression. Amphetamine 155-166 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 16084549-4 2006 Results showed that AMPH treatment decreased food intake, which was correlated with changes of NPY mRNA level, but increased c-fos, c-jun and superoxide dismutase (SOD) mRNA levels in hypothalamus. Amphetamine 20-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 16084549-7 2006 It was suggested that c-fos/c-jun signaling might involve in the central regulation of AMPH-mediated feeding suppression via the modulation of NPY gene expression. Amphetamine 87-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 16498678-7 2006 In addition, CTb/Fos double-labeled cells constituted 43% of all the singly CTb-labeled cells counted in the DPGi compared with 29% for the LPGi, 18% for the rostral PAG, and 10% or less for the other structures. phenylacetylglycine 166-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 16623859-0 2006 Assessment by c-Fos immunostaining of changes in brain neural activity induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and leptin in rats. Polychlorinated Dibenzodioxins 82-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 17152352-0 2006 Expression of the c-Fos gene in hypothalamic cells and cytotoxic activity of natural killer cells in the spleen of rats after treatment with Cytoxan. Cyclophosphamide 141-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 16623859-0 2006 Assessment by c-Fos immunostaining of changes in brain neural activity induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and leptin in rats. Polychlorinated Dibenzodioxins 119-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 17152352-2 2006 Administration of Cytoxan in a dose of 60 mg/kg increased the number of c-Fos-positive cells in the ventromedial hypothalamus and lateral hypothalamic area and reduced interferon-alpha-induced cytotoxic activity of natural killer cells. Cyclophosphamide 18-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 16344929-5 2006 Furthermore, expression of Fos protein in the lumbar dorsal horn was immunohistochemically investigated following the injection of formalin into the hindpaw during TMJ inflammation. Formaldehyde 131-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 16242920-10 2006 In situ hybridisation studies indicated significant increases in b-ZIP transcription factors (CREM, ICER, CBP, and c-fos) elicited by MK-801 and decreases in c-fos elicited by cocaine. Dizocilpine Maleate 134-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 16344929-8 2006 The number of Fos-positive neurons in the lumbar dorsal horn ipsilateral to the formalin injection at 1 and 7 days after CFA injection into the TMJ were similar to those in the non-CFA group; however, those were significantly increased in the laminae I-II and V-VI of the lumbar dorsal horn at 14 days after CFA injection. Formaldehyde 80-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 16324724-3 2006 In the present studies, the selective dopamine D4 agonist PD168077 induces c-Fos expression and extracellular signal regulated kinase (ERK) phosphorylation in the hypothalamic paraventricular nucleus (PVN), a site known to regulate proerectile activity. Dopamine 38-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 16623831-8 2006 Mirtazapine increased the number of c-Fos-positive nuclei in the central amygdala and dentate gyrus. Mirtazapine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 16324724-3 2006 In the present studies, the selective dopamine D4 agonist PD168077 induces c-Fos expression and extracellular signal regulated kinase (ERK) phosphorylation in the hypothalamic paraventricular nucleus (PVN), a site known to regulate proerectile activity. N-((4-(2-cyanophenyl)-1-piperazinyl)methyl)-3-methylbenzamide 58-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 16324724-4 2006 The selective dopamine D4 receptor antagonist A-381393 blocked both c-Fos expression and ERK1/2 phosphorylation produced by PD168077. 2-(4-(3,4-dimethylphenyl)piperazin-1-ylmethyl)-1H-benzimidazole 46-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 16427151-5 2006 We also found that lithium chloride (LiCl, 0.5 and l.0 mEq), a compound known to activate oxytocinergic neurons, also significantly increased the percentage of c-Fos positive oxytocin neurons in all PVN portions. Lithium Chloride 19-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 16427151-5 2006 We also found that lithium chloride (LiCl, 0.5 and l.0 mEq), a compound known to activate oxytocinergic neurons, also significantly increased the percentage of c-Fos positive oxytocin neurons in all PVN portions. Lithium Chloride 37-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 16324724-7 2006 Dopamine D4 receptor and c-Fos coexpression in the PVN was observed using double immunohistochemical labeling, suggesting that PD168077-induced signaling may result from direct dopamine D4 receptor activation. N-((4-(2-cyanophenyl)-1-piperazinyl)methyl)-3-methylbenzamide 127-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 16324724-9 2006 Further, the ability of the selective dopamine D4 antagonist A-381393 alone to reduce c-Fos expression below control levels may imply the presence of a tonic dopamine D4 receptor activation under basal conditions in vivo. dopamine-d4 38-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 16324724-9 2006 Further, the ability of the selective dopamine D4 antagonist A-381393 alone to reduce c-Fos expression below control levels may imply the presence of a tonic dopamine D4 receptor activation under basal conditions in vivo. 2-(4-(3,4-dimethylphenyl)piperazin-1-ylmethyl)-1H-benzimidazole 61-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 16521035-0 2006 5-HT receptor subtypes involved in the anxiogenic-like action and associated Fos response of acute fluoxetine treatment in rats. Fluoxetine 99-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-80 16470400-0 2006 Previous experience of ethanol withdrawal increases withdrawal-induced c-fos expression in limbic areas, but not withdrawal-induced anxiety and prevents withdrawal-induced elevations in plasma corticosterone. Ethanol 23-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 16494863-2 2006 The effect of citalopram on CFS-induced c-Fos expression was investigated using immunohistochemistry. Citalopram 14-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 16494863-2 2006 The effect of citalopram on CFS-induced c-Fos expression was investigated using immunohistochemistry. thallium sulfate 28-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 16494863-3 2006 Systemic administration of citalopram attenuated contextual CFS-induced c-Fos expression in the secondary motor cortex, primary somatosensory cortex, and basolateral nucleus of the amygdala. Citalopram 27-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 16448624-9 2006 However, berberine significantly attenuated MEK/ERK activation and downstream target (Egr-1, c-Fos, Cyclin D1 and PDGF-A) expression after mechanic injury in vitro. Berberine 9-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 16545369-4 2006 Nicotine increased the percentage of orexin neurons expressing Fos without a significant effect on non-orexin neurons. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 16521035-4 2006 RESULTS: Acute fluoxetine increased the airjet-induced escape behaviour and Fos expression in the LC of saline-pretreated rats. Fluoxetine 15-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 16521035-4 2006 RESULTS: Acute fluoxetine increased the airjet-induced escape behaviour and Fos expression in the LC of saline-pretreated rats. Sodium Chloride 104-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 16521035-7 2006 Chronic treatment with fluoxetine abolished the anxiogenic-like effect and led to a normalization of the enhanced fluoxetine-induced Fos response to airjet. Fluoxetine 23-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-136 16521035-7 2006 Chronic treatment with fluoxetine abolished the anxiogenic-like effect and led to a normalization of the enhanced fluoxetine-induced Fos response to airjet. Fluoxetine 114-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-136 16427032-4 2006 The enhancement in the number of c-Fos-positive cells in the L4-5 spinal dorsal horn (DH) and the magnitude of paw edema induced by formalin injection during phase II were significantly reduced by minocycline. Formaldehyde 132-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 16492833-0 2006 Licking and c-fos expression in the dorsal horn of the spinal cord after the formalin test. Formaldehyde 77-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 16492833-1 2006 We investigated whether c-fos expression in the dorsal horn is affected by licking in the formalin test. Formaldehyde 90-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 16492833-8 2006 The results indicated that the licking action increased c-fos expression of the lumbar dorsal horn in the formalin test. Formaldehyde 106-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 16168987-4 2006 During 7 days after the intraperitoneal injection of MNU (60 mg/kg), rat retinas exhibited DNA fragmentation characteristic of apoptosis and activation of PARP, phosphorylation of JNK and c-Jun, induction of AP-1 (c-Jun and c-Fos) and Bax, as well as photoreceptor cell loss. Methylnitrosourea 53-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 224-229 16426583-7 2006 Rivastigmine significantly increased c-Fos immunoreactivity in medial prefrontal cortex and the hippocampus, but not in the septum and dorsal raphe nucleus. Rivastigmine 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 16426583-8 2006 5-HT depletion decreased ACh-induced c-Fos immunoreactivity in the dentate gyrus. Acetylcholine 25-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 16453312-3 2006 In DPSPX-treated animals, a 20% increase in blood pressure was achieved along with a decrease in Fos expression in the superficial (laminae I-II) and deep (laminae III-VII) dorsal horn. 1,3-dipropyl-8-(4-sulfophenyl)xanthine 3-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 16453312-5 2006 Lesioning the VLMlat with quinolinic acid prevented the decrease in Fos expression at the spinal cord of DPSPX-hypertensive rats whereas in normotensive animals, no changes in Fos expression were detected. Quinolinic Acid 26-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 16453312-5 2006 Lesioning the VLMlat with quinolinic acid prevented the decrease in Fos expression at the spinal cord of DPSPX-hypertensive rats whereas in normotensive animals, no changes in Fos expression were detected. 1,3-dipropyl-8-(4-sulfophenyl)xanthine 105-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 16309718-0 2006 Unique patterns of FOS, phospho-CREB and BrdU immunoreactivity in the female rat brain following chronic stress and citalopram treatment. Citalopram 116-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 16677576-7 2006 The c-fos protein expression of the VSMCs after thrombin stimulation for 24 h increased by 156.0% +/- 11.3% (P < 0.05), and the bcl-2 protein expression of the VSMCs pretreated with puerarin of the concentrations of 1.5 x 10(-5), 1.5 x 10(-4), and 1.5 x 10(-3) mol/L, and then stimulated by thrombin was significantly lower than that of the VSMCs only stimulated by thrombin with the suppression rates of 20.7% +/- 2.1%, 31.6% +/- 5.2%, and 44.5% +/- 7.5% respectively (all P < 0.05). puerarin 185-193 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 16677576-12 2006 The inhibitory effect of puerarin is closely related with the suppression of the protein expression of c-fos and bcl-2n, and partly related with the suppression of the TR mRNA expression. puerarin 25-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 16385558-8 2006 In conclusion, our previously reported decreases in c-Fos and PKA expression in the NTS following pretreatment with low doses of naltrexone may be partially explained by a greater inhibition of NTS neurons resulting from increased muOR expression in this region. Naltrexone 129-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 16427032-4 2006 The enhancement in the number of c-Fos-positive cells in the L4-5 spinal dorsal horn (DH) and the magnitude of paw edema induced by formalin injection during phase II were significantly reduced by minocycline. Minocycline 197-208 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 16492117-5 2006 In the latter, Fos expression upon SD was enhanced in the amygdala and hypothalamic areas compared with LABs, whereas it was diminished in prefrontal and brainstem areas. SD 0006 35-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-18 16673821-4 2006 RESULTS: The expression of c-FOS protein in cortex and hippocampus increased significantly, the accumulation of mercury in the brain induced by 0.05 mg/Kg MMC for 20 min had no significant difference compared with the control group. methylmercuric chloride 155-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 16438960-8 2006 Finally, systemic morphine reduced post-operative pain and Fos-like immunoreactivity in a naloxone reversible manner, with greater potency and efficacy on behavioral endpoints than on Fos-like immunoreactivity. Morphine 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 16438960-8 2006 Finally, systemic morphine reduced post-operative pain and Fos-like immunoreactivity in a naloxone reversible manner, with greater potency and efficacy on behavioral endpoints than on Fos-like immunoreactivity. Morphine 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 184-187 16438960-8 2006 Finally, systemic morphine reduced post-operative pain and Fos-like immunoreactivity in a naloxone reversible manner, with greater potency and efficacy on behavioral endpoints than on Fos-like immunoreactivity. Naloxone 90-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 16492237-8 2006 c-fos experiments: Compared with vehicle, all doses of parecoxib (1 mg/kg, 10 mg/kg, 50 mg/kg) significantly reduced the number of c-fos positive cells in the ipsilateral TNC (P < .05). parecoxib 55-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 16492237-8 2006 c-fos experiments: Compared with vehicle, all doses of parecoxib (1 mg/kg, 10 mg/kg, 50 mg/kg) significantly reduced the number of c-fos positive cells in the ipsilateral TNC (P < .05). parecoxib 55-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-136 16492237-9 2006 The number of c-fos positive cells in the ipsilateral TNC was 50 +/- 2.7 (mean +/- SEM) under control conditions and 9.1 +/- 0.6 after pretreatment with 50 mg/kg parecoxib. parecoxib 162-171 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 16575111-0 2006 Changes in the expression of c-fos & heat shock protein genes & blood flow velocity in the brain of rats undergoing myocardial ischaemia/reperfusion. Adenosine Monophosphate 67-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 16452668-9 2006 Moreover, DHT, 3beta-diol, and diarylpropionitrile treatment significantly decreased restraint-induced c-fos mRNA expression in the PVN. Dihydrotestosterone 10-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 16452668-9 2006 Moreover, DHT, 3beta-diol, and diarylpropionitrile treatment significantly decreased restraint-induced c-fos mRNA expression in the PVN. 3beta-diol 15-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 16452668-9 2006 Moreover, DHT, 3beta-diol, and diarylpropionitrile treatment significantly decreased restraint-induced c-fos mRNA expression in the PVN. diarylpropionitrile 31-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 16360647-8 2006 In addition, riluzole appeared to produce nonspecific effects on c-Fos expression by itself, without specifically modifying c-Fos expression following naloxone-precipitated withdrawal in any region of the amygdala examined, suggesting that the amygdala may not serve as a specific site of action for riluzole. Riluzole 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 16386717-3 2006 In the present experiment, an immunohistochemical analysis of the immediate early protein c-Fos was performed as a marker for cellular activity in the brains of suspended rat pups treated with l-DOPA at P15 and P25. Levodopa 193-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 16386717-5 2006 Only P15 rat pups injected with L-DOPA engaged in air stepping and expressed the highest levels of c-Fos reactivity in output nuclei of the basal ganglia, as well as the pedunculopontine (PPN) and cuneiform (Cnf) nuclei. Levodopa 32-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 17007911-0 2006 Alterations of seizure-induced c-fos immunolabelling and gene expression in the rat cerebral cortex following dexamethasone treatment. Dexamethasone 110-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 17007911-1 2006 We examined the effects of dexamethasone on the expression of the inducible transcription factor c-fos in 4-aminopyridine (4-AP) seizures. Dexamethasone 27-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 17007911-1 2006 We examined the effects of dexamethasone on the expression of the inducible transcription factor c-fos in 4-aminopyridine (4-AP) seizures. 4-Aminopyridine 106-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 17007911-1 2006 We examined the effects of dexamethasone on the expression of the inducible transcription factor c-fos in 4-aminopyridine (4-AP) seizures. 4-Aminopyridine 123-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 17007911-2 2006 Induction of c-fos mRNA due to 4-AP-elicited convulsion was detected by means of the polymerase chain reaction (PCR) in samples from the neocortex. 4-Aminopyridine 31-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 17007911-8 2006 Pretreatment with dexamethasone resulted in a dose-dependent, significant decrease of seizure-induced Fos-protein immunoreactivity in the neocortex, in the hilum of the dentate fascia, as well as in regions CA1-3 of the hippocampus, compared to control animals. Dexamethasone 18-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 17007911-9 2006 Brains processed for mRNA isolation and PCR, displayed a significant increase of c-fos mRNA following the 4-AP treatment, while pretreatment with dexamethasone did not prevent or decrease this boosted c-fos mRNA expression. 4-Aminopyridine 106-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 17007911-10 2006 We conclude that seizure-induced c-fos expression and intracellular Fos-protein localization are mediated by transmitter and receptor systems, and dexamethasone significantly decreases Fos immunoreactivity, probably by regulating the intracellular traffic of the protein. Dexamethasone 147-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 185-188 17163588-9 2006 Its Fos-suppressive effects were mostly blocked by naloxone, an opioid antagonist. Naloxone 51-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-7 17163588-10 2006 In addition, acupuncture at St36 or Li4 significantly decreased Fos-containing PNMT, and this effect was also reversed by naloxone. Naloxone 122-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 16687308-3 2006 At the beginning of the night, norepinephrine (NE) elicits a rapid and sustained phosphorylation of CREB into pCREB and a transient synthesis of the immediate early gene products c-FOS and c-JUN that peak 3 h after dark onset. Norepinephrine 31-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-184 16687308-5 2006 Interestingly, injection of the protein synthesis inhibitor cycloheximide before, but not after, the c-FOS/c-JUN peak markedly reduces Aanat mRNA levels. Cycloheximide 60-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 17333617-6 2006 About 50 % of double-labeled (Fos-ir and NADPH-diaphorase reactive) cells were found in Pa nucleus. Protactinium 88-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 16483831-0 2006 1alpha,25(OH)(2)-Vitamin D(3) stimulates intestinal cell p38 MAPK activity and increases c-Fos expression. 1alpha 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 16483831-0 2006 1alpha,25(OH)(2)-Vitamin D(3) stimulates intestinal cell p38 MAPK activity and increases c-Fos expression. 25(oh)(2)-vitamin d 7-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 16483831-8 2006 Incubation of intestinal cells with the hormone was followed by a rapid induction of c-Fos expression which was blocked by SB 203580 and partially suppressed by the ERK1/2 inhibitor PD 98059. SB 203580 123-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 16483831-8 2006 Incubation of intestinal cells with the hormone was followed by a rapid induction of c-Fos expression which was blocked by SB 203580 and partially suppressed by the ERK1/2 inhibitor PD 98059. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 182-190 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 17078562-0 2006 Expression of c-fos and oxidative stress on brain of rats reared on food from mercury-selenium coexisting mining area. Mercury 78-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 17078562-0 2006 Expression of c-fos and oxidative stress on brain of rats reared on food from mercury-selenium coexisting mining area. Selenium 86-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 17078562-2 2006 In order to study the neurotoxicity of food from Wanshan mercury mine area and probe into the effect of food from Wanshan mercury miner area on the changes of brain oxidative damage and expression of c-fos gene. Mercury 122-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 200-205 17078562-4 2006 The results showed the levels of GSH, MDA, SOD and of GSH-dependent enzymes in the rat brain changed between exposure groups and control group; The mercury polluted rice induced significantly the expression of c-fos mRNA; the c-FOS positive cells in hippocampus and cortex of exposure groups were significant different from control group (P<0.01). Mercury 148-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 210-215 17078562-4 2006 The results showed the levels of GSH, MDA, SOD and of GSH-dependent enzymes in the rat brain changed between exposure groups and control group; The mercury polluted rice induced significantly the expression of c-fos mRNA; the c-FOS positive cells in hippocampus and cortex of exposure groups were significant different from control group (P<0.01). Mercury 148-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 226-231 17078562-5 2006 It could be concluded that oxidative stress signals could contribute to the induction of immediate early genes (IEGs); free radicals and their by-products might not only cause oxidative damage, but also influenced gene expression; IEGs c-fos participated in the toxicity process of brain injury by mercury polluted food. Mercury 298-305 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 236-241 16372144-0 2006 Midazolam inhibits neophobia-induced Fos expression in the rat hippocampus. Midazolam 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 16372144-1 2006 The effect of midazolam on expression of c-Fos protein was examined in the rat hippocampus, following the open field test of neophobia. Midazolam 14-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 16372144-2 2006 It was found that pretreatment of rats with midazolam, at the dose of 0.5 mg/kg, enhanced rat exploratory behavior, and inhibited neophobia related stimulation of c-Fos in the CA-1 and CA-3 areas of the hippocampus. Midazolam 44-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-168 17314472-9 2006 Oil-implanted OVX rats showed significantly diminished numbers of Fos-ir-positive neurons in each neural structure after repeated hypoglycemia. Oils 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 15920503-0 2006 Perinatal exposure to delta(9)-tetrahydrocannabinol alters heroin-induced place conditioning and fos-immunoreactivity. Dronabinol 22-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 15920503-9 2006 Perinatal THC exposure significantly increased heroin-induced Fos-IR in the dorsomedial CPu. Dronabinol 10-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 15920503-9 2006 Perinatal THC exposure significantly increased heroin-induced Fos-IR in the dorsomedial CPu. Heroin 47-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 15920503-10 2006 Conversely, perinatal THC exposure reduced heroin-induced Fos-IR in the NAC (shell), BNST, CEA, dorsolateral and lateral PAG, VTA, and EW. Dronabinol 22-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 15920503-10 2006 Conversely, perinatal THC exposure reduced heroin-induced Fos-IR in the NAC (shell), BNST, CEA, dorsolateral and lateral PAG, VTA, and EW. Heroin 43-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 16359808-0 2006 Effects of environmental and pharmacological stressors on c-fos and corticotropin-releasing factor mRNA in rat brain: Relationship to the reinstatement of alcohol seeking. Alcohols 155-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 16359808-6 2006 Exposure of rats to the environmental stressors footshock, restraint or social defeat, or the pharmacological stressors yohimbine or FG-7142 increased levels of the mRNAs for c-fos and CRF in the brain in a number of areas previously shown to be responsive to stressors. Yohimbine 120-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 175-180 16359808-7 2006 We found regionally specific effects of the stressors on c-fos and CRF mRNA in brain regions associated with the rewarding effects of alcohol and other abused drugs. Alcohols 134-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 16359808-8 2006 The two stressors we have previously shown to be effective in inducing reinstatement of alcohol seeking, footshock and yohimbine, induced c-fos mRNA in the shell of the nucleus accumbens, and the basolateral and central amygdalar nuclei. Alcohols 88-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 16359808-8 2006 The two stressors we have previously shown to be effective in inducing reinstatement of alcohol seeking, footshock and yohimbine, induced c-fos mRNA in the shell of the nucleus accumbens, and the basolateral and central amygdalar nuclei. Yohimbine 119-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 16488545-0 2006 Injections of urocortin 1 into the basolateral amygdala induce anxiety-like behavior and c-Fos expression in brainstem serotonergic neurons. Urocortins 14-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 16531639-0 2006 Effects of methadone and morphine on c-Fos expression in the rat brain: similarities and differences. Methadone 11-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 16531639-0 2006 Effects of methadone and morphine on c-Fos expression in the rat brain: similarities and differences. Morphine 25-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 16531639-1 2006 This is the first study designed to compare the pattern of stimulation of c-Fos in selected brain structures after an acute administration of morphine and methadone. Morphine 142-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 16531639-1 2006 This is the first study designed to compare the pattern of stimulation of c-Fos in selected brain structures after an acute administration of morphine and methadone. Methadone 155-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 16531639-2 2006 Methadone and morphine induced activation of c-Fos protein in the terminal forebrain projecting areas of the brain dopaminergic system, i.e. the striatum and nucleus accumbens. Methadone 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 16531639-2 2006 Methadone and morphine induced activation of c-Fos protein in the terminal forebrain projecting areas of the brain dopaminergic system, i.e. the striatum and nucleus accumbens. Morphine 14-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 16328378-0 2006 The effects of ziprasidone on regional c-Fos expression in the rat forebrain. ziprasidone 15-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 16328378-2 2006 OBJECTIVE: To use c-Fos immunohistochemistry to investigate the functional neuroanatomical profile of the newly introduced atypical agent ziprasidone. ziprasidone 138-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 16328378-3 2006 MATERIALS AND METHODS: c-Fos immunohistochemistry was performed on paraformaldehyde-fixed cryosections of rat brains obtained, initially, from animals 2, 4, or 6 h after oral administration of 10 mg/kg ziprasidone or vehicle and, subsequently, from animals 2 h after oral administration of 1, 3, or 10 mg/kg ziprasidone or vehicle. paraform 67-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 16328378-5 2006 RESULTS: Ziprasidone induced a time-dependent increase in the density of c-Fos-positive nuclei that was maximal at 2 h. At the 2 h time-point, c-Fos expression was significantly (p<0.05) elevated in the shell and core of the nucleus accumbens, lateral and medial caudate putamen, and lateral septum. ziprasidone 9-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 16328378-5 2006 RESULTS: Ziprasidone induced a time-dependent increase in the density of c-Fos-positive nuclei that was maximal at 2 h. At the 2 h time-point, c-Fos expression was significantly (p<0.05) elevated in the shell and core of the nucleus accumbens, lateral and medial caudate putamen, and lateral septum. ziprasidone 9-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-148 16328378-7 2006 Ziprasidone-induced c-Fos expression was dose-dependent with significant (p<0.05) c-Fos expression observed in the nucleus accumbens (shell and core) and caudate putamen (lateral and medial) at 3 and 10 mg/kg and in the lateral septum at 10 mg/kg. ziprasidone 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 16328378-7 2006 Ziprasidone-induced c-Fos expression was dose-dependent with significant (p<0.05) c-Fos expression observed in the nucleus accumbens (shell and core) and caudate putamen (lateral and medial) at 3 and 10 mg/kg and in the lateral septum at 10 mg/kg. ziprasidone 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 16328378-9 2006 Thus, the observed pattern of c-Fos expression induced in rat brain by ziprasidone is consistent with its reported clinical effects, namely, efficacy against positive symptoms with a therapeutic window over motor side effects and with some activity against negative symptoms. ziprasidone 71-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 16304628-7 2005 Lesions of Vc with a soma-selective neurotoxin, ibotenic acid, significantly reduced inflammation-induced Fos expression as well as the number of FluoroGold/Fos double-labeled neurons in the ventral Vi/Vc (P<0.05). Ibotenic Acid 48-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 16304628-7 2005 Lesions of Vc with a soma-selective neurotoxin, ibotenic acid, significantly reduced inflammation-induced Fos expression as well as the number of FluoroGold/Fos double-labeled neurons in the ventral Vi/Vc (P<0.05). Ibotenic Acid 48-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-160 16324697-0 2005 The atypical dopamine D1 receptor agonist SKF 83959 induces striatal Fos expression in rats. SK and F 83959 42-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 16324697-3 2005 The current report demonstrates that SKF 83959 induces pronounced, nonpatchy, expression of the immediate-early gene product Fos in the striatum of intact rats which can be converted to a patchy pattern by pretreatment with the dopamine D2-like receptor agonist quinpirole. SK and F 83959 37-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-128 16324697-3 2005 The current report demonstrates that SKF 83959 induces pronounced, nonpatchy, expression of the immediate-early gene product Fos in the striatum of intact rats which can be converted to a patchy pattern by pretreatment with the dopamine D2-like receptor agonist quinpirole. Dopamine 228-236 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-128 16324697-3 2005 The current report demonstrates that SKF 83959 induces pronounced, nonpatchy, expression of the immediate-early gene product Fos in the striatum of intact rats which can be converted to a patchy pattern by pretreatment with the dopamine D2-like receptor agonist quinpirole. Quinpirole 262-272 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-128 16105718-11 2005 administration of CCK-8 is a more potent stimulant of Fos-LI in the neurons of the myenteric plexus of the duodenum and jejunum than i.v. cholecystokinin 8 18-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 16289800-6 2005 The spinal ERK inhibition or knockdown also reduced morphine withdrawal-induced phosphorylation of cAMP response element binding protein (CREB), which is one of the important downstream substrates of ERK pathway, and Fos expression. Morphine 52-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 217-220 16095819-0 2005 Buspirone attenuates conditioned fear-induced c-Fos expression in the rat hippocampus. Buspirone 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 16410675-6 2006 In the CsA-treated animals C-FOS expression was found to increase, but the expression level reduced to a statistically insignificant level within 48 h after the ischemia. Cyclosporine 7-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 16095819-1 2005 The role of hippocampus in the anxiolytic-like effect of buspirone in the conditioned emotional response test (CER, a freezing response), was examined by immunocytochemical detection of the c-Fos protein. Buspirone 57-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 190-195 16095819-5 2005 Pretreatment with buspirone (1.5 mg/kg) significantly attenuated the effects of aversive memory on c-Fos protein expression in the CA-1 and CA-3 layers of the hippocampus. Buspirone 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 16190878-0 2005 Role of PKC-alpha,gamma isoforms in regulation of c-Fos and TH expression after naloxone-induced morphine withdrawal in the hypothalamic PVN and medulla oblongata catecholaminergic cell groups. Morphine 97-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 16219028-1 2005 Repeated cocaine administration to rats outside their home cages sensitizes the behavioral effects of the drug, and enhances induction of the immediate early gene product Fos in nucleus accumbens. Cocaine 9-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-174 16219028-2 2005 We hypothesized that the same treatment regimen would also enhance cocaine-induced activation of intracellular signaling kinases that phosphorylate cyclic AMP-regulated element-binding protein (CREB), an important mediator of c-fos transcription. Cocaine 67-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 226-231 16165275-3 2005 Furthermore, we have shown that manumycin increased the activity of c-Fos in the M-CR3B cells and decreased the activity of NF-kappaB, while l-NAME decreased the activities of both transcription factors, and accelerated apoptosis of M-CR3B cells. manumycin 32-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 16150912-8 2005 In the PVN, c-fos mRNA was suppressed by the highest dose of dex, but this effect showed a degree of resistance after long-term oral treatment. Dexamethasone 61-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 16150912-9 2005 c-fos mRNA responses in the anterior pituitary followed those in PVN and reflect central drive of the HPA axis even if corticosterone responses are strongly reduced. Corticosterone 119-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 16190878-0 2005 Role of PKC-alpha,gamma isoforms in regulation of c-Fos and TH expression after naloxone-induced morphine withdrawal in the hypothalamic PVN and medulla oblongata catecholaminergic cell groups. Naloxone 80-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 16190878-1 2005 We previously demonstrated that morphine withdrawal induced hyperactivity of the hypothalamus-pituitary-adrenocortical axis by activation of noradrenergic pathways innervating the hypothalamic paraventricular nucleus (PVN), as evaluated by Fos expression and corticosterone release. Morphine 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 240-243 16190878-6 2005 Morphine withdrawal induced c-Fos expression in the PVN and NTS/VLM, indicating an activation of neurons in those nuclei. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 16190878-8 2005 Infusion of calphostin C, a selective protein kinase C inhibitor, produced a reduction in the morphine withdrawal-induced c-Fos expression. calphostin C 12-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 16190878-8 2005 Infusion of calphostin C, a selective protein kinase C inhibitor, produced a reduction in the morphine withdrawal-induced c-Fos expression. Morphine 94-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 16040065-3 2005 This was assessed by immunohistochemical analysis of cFos expression in the rat brains after retro-dialysis application of NMDA (50mM, 10 min) into VH, in absence and in presence of either the D1/D5 receptor antagonist SCH 23390 (100 and 250 microM, 60 min) or the D2 receptor antagonist raclopride (100 and 250 microM, 60 min). N-Methylaspartate 123-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-57 16210350-0 2005 Different neuronal populations of the rat median preoptic nucleus express c-fos during sleep and in response to hypertonic saline or angiotensin-II. Sodium Chloride 123-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 16040065-4 2005 NMDA induced a robust increase in the cFos expression in the NAc shell, both in the ipsilateral and contralateral side. N-Methylaspartate 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-42 16040065-6 2005 Simultaneous application of SCH 23390 and NMDA into the VH attenuated the NMDA-evoked cFos expression in NAc shell. SCH 23390 28-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-90 16040065-6 2005 Simultaneous application of SCH 23390 and NMDA into the VH attenuated the NMDA-evoked cFos expression in NAc shell. N-Methylaspartate 42-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-90 16040065-6 2005 Simultaneous application of SCH 23390 and NMDA into the VH attenuated the NMDA-evoked cFos expression in NAc shell. N-Methylaspartate 74-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-90 16228990-8 2005 Retrograde neural tracing demonstrated that neurons activated by YO at 5.0 mg/kg BW included medullary and pontine neurons that project to the central nucleus of the amygdala and to the lateral bed nucleus of the stria terminalis, the latter region receiving comparatively greater input by Fos-positive neurons. Yohimbine 65-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 290-293 16137723-0 2005 Repeated exposure to Delta(9)-tetrahydrocannabinol alters heroin-induced locomotor sensitisation and Fos-immunoreactivity. Dronabinol 21-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-104 16137723-11 2005 Acute heroin increased Fos-IR in drug-naive rats in the caudate-putamen (CPu; central, medial and dorsomedial regions), nucleus accumbens (NAC; core and shell regions), bed nucleus of the stria terminalis (BNST), lateral septum, central nucleus of the amygdala (CEA), periaqueductal grey (PAG; dorsolateral, dorsomedial, and lateral), and the Edinger-Westphal nucleus. Heroin 6-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 16137723-12 2005 Pre-exposure to THC significantly increased heroin-induced Fos-IR in the dorsomedial CPu and the NAC (core). Dronabinol 16-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 16137723-12 2005 Pre-exposure to THC significantly increased heroin-induced Fos-IR in the dorsomedial CPu and the NAC (core). Heroin 44-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 16137723-13 2005 Conversely, THC pre-exposure reduced heroin-induced Fos-IR in the BNST, CEA, and the PAG (dorsolateral and lateral). Dronabinol 12-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 16137723-13 2005 Conversely, THC pre-exposure reduced heroin-induced Fos-IR in the BNST, CEA, and the PAG (dorsolateral and lateral). Heroin 37-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 15978629-6 2005 c-fos expression was used as a marker of neuronal activation and revealed by immunohistochemistry 1h after intraperitoneal acetic acid injection and 2 h after colonic inflammation. Hydrogen 98-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 16212993-0 2005 Losartan blocks drinking and cFos expression induced by central ornithine vasotocin in rats. Losartan 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-33 16212993-0 2005 Losartan blocks drinking and cFos expression induced by central ornithine vasotocin in rats. ornithine vasotocin 64-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-33 16223404-6 2005 RESULTS: There were four immediate early genes/transcription factors (early growth response 1, c-fos, nerve growth factor-induced factor A and Knox-24) whose mRNA expression was increased to more than 1.4-fold of control levels under the conditions of isoflurane, OGD or isoflurane plus OGD. Isoflurane 252-262 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 15985455-3 2005 The numbers of Fos immunoreactive (IR) positive neurons in the DMV, NTS, and AP were low in euthyroid rats but significantly higher in the 4-wk duration in hypothyroid rats, which were prevented by simultaneous T4 replacement. (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 63-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-18 15985455-5 2005 There were significant negative correlations between T4 levels and the numbers of Fos-IR-positive neurons in the DMV (r = -0.6388, P < 0.008), NTS (r = -0.6741, P < 0.003), and AP (r = -0.5622, P < 0.004). (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 113-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 15985455-6 2005 Double staining showed that Fos immunoreactivity in the DMV of hypothyroid rats was mostly localized in choline acetyltransferase-containing neurons. (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 56-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 16394142-12 2005 Antisense RIalpha in the liver induced the phase II enzymes, glutathione S-transferase and quinone oxidoreductase, c-fos protein, and activator protein-1 (AP-1)- and cAMP response element (CRE)-directed transcription. rialpha 10-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 15978629-6 2005 c-fos expression was used as a marker of neuronal activation and revealed by immunohistochemistry 1h after intraperitoneal acetic acid injection and 2 h after colonic inflammation. Acetic Acid 123-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 15978629-7 2005 Nor-trimebutine decreased Fos expression in the thoraco-lumbar (peritoneal irritation) and lumbo-sacral (colonic inflammation) spinal cord in laminae I, IIo V, VII and X. N-desmethyltrimebutine 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 16175568-5 2005 Understanding the activation of immediate-early genes such as c-fos or ICER in response to a single LiCl injection is an important first step in understanding the long-term changes in gene expression elicited by lithium that are involved in its therapeutic and toxic effect. Lithium Chloride 100-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 16175568-5 2005 Understanding the activation of immediate-early genes such as c-fos or ICER in response to a single LiCl injection is an important first step in understanding the long-term changes in gene expression elicited by lithium that are involved in its therapeutic and toxic effect. Lithium 212-219 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 16153612-0 2005 Muscarinic receptor blockade attenuates reserpine-mediated Fos induction in the rat striatopallidal pathway. Reserpine 40-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 16199019-1 2005 In the present study, a semi-quantitative analysis of Fos expression by mustard oil was performed. mustard oil 72-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 16199019-5 2005 The number of Fos-labelled cells gradually increased in a dose dependent manner in both sides of superficial layers (laminae I-II) of the spinal cord with increasing concentration of mustard oil. mustard oil 183-194 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 16061485-9 2005 In addition, cotransfection experiments indicated that the c-Fos/c-Jun heterodimer is responsible for cAMP-dependent Lot1 transcriptional activation. Cyclic AMP 102-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 16061485-10 2005 In conclusion, our data demonstrate that, in CGC, Lot1 is under the transcriptional control of cAMP through an AP1 site regulated by the c-Fos/c-Jun heterodimer and suggest that this gene may be an important element of the cAMP-mediated pathway that regulates neuronal proliferation through the protein kinase A-MEK signaling cascade. Cyclic AMP 95-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 16061485-10 2005 In conclusion, our data demonstrate that, in CGC, Lot1 is under the transcriptional control of cAMP through an AP1 site regulated by the c-Fos/c-Jun heterodimer and suggest that this gene may be an important element of the cAMP-mediated pathway that regulates neuronal proliferation through the protein kinase A-MEK signaling cascade. Cyclic AMP 223-227 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 16055088-6 2005 Pre-treatment as well as post-treatment of rats with a JNK inhibitor, SP600125, significantly attenuated thermal hyperalgesia, as assessed by paw-withdrawal latency, and the upregulation of c-fos immunoreactivity in dorsal horn neurons. pyrazolanthrone 70-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 190-195 16098547-1 2005 Intestinal infusion of nutrients, such as glucose and oleic acid, increase Fos-like immunoreactivity (Fos-LI) in both the enteric nervous system and neurons of the dorsal vagal complex (DVC) of the hindbrain. Glucose 42-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 16098547-1 2005 Intestinal infusion of nutrients, such as glucose and oleic acid, increase Fos-like immunoreactivity (Fos-LI) in both the enteric nervous system and neurons of the dorsal vagal complex (DVC) of the hindbrain. Glucose 42-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 16098547-1 2005 Intestinal infusion of nutrients, such as glucose and oleic acid, increase Fos-like immunoreactivity (Fos-LI) in both the enteric nervous system and neurons of the dorsal vagal complex (DVC) of the hindbrain. Oleic Acid 54-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 16098547-1 2005 Intestinal infusion of nutrients, such as glucose and oleic acid, increase Fos-like immunoreactivity (Fos-LI) in both the enteric nervous system and neurons of the dorsal vagal complex (DVC) of the hindbrain. Oleic Acid 54-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 16098547-3 2005 Both oleate and glucose infusions increased Fos-LI in the DVC. Oleic Acid 5-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 16098547-3 2005 Both oleate and glucose infusions increased Fos-LI in the DVC. Glucose 16-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 16326368-7 2005 Capsaicin-evoked Fos protein expression was increased in the superficial and neck regions of the dorsal horn of adult P0-CFA-treated rats relative to P0-vehicle-treated rats. Capsaicin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 16300424-0 2005 Relationship of cocaine-induced c-Fos expression to behaviors and the role of serotonin 5-HT2A receptors in cocaine-induced c-Fos expression. Cocaine 16-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 16300424-0 2005 Relationship of cocaine-induced c-Fos expression to behaviors and the role of serotonin 5-HT2A receptors in cocaine-induced c-Fos expression. Cocaine 108-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 16300424-2 2005 The present study used the expression of c-Fos protein as a marker to identify brain areas through which 5-HT2A receptors may modulate cocaine-induced behaviors. Cocaine 135-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 16300424-3 2005 Significant correlations were observed between cocaine-induced hyperactivity and c-Fos expression in the nucleus accumbens core (NAcC), caudate-putamen (CPu), and subthalamic nucleus. Cocaine 47-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 16300424-3 2005 Significant correlations were observed between cocaine-induced hyperactivity and c-Fos expression in the nucleus accumbens core (NAcC), caudate-putamen (CPu), and subthalamic nucleus. nacc 129-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 16300424-4 2005 In a separate experiment, a low, behaviorally relevant dose of cocaine was found to increase c-Fos immunoreactivity in the medial CPu, NAcC, and nucleus accumbens shell (NAcSh). Cocaine 63-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 16300424-4 2005 In a separate experiment, a low, behaviorally relevant dose of cocaine was found to increase c-Fos immunoreactivity in the medial CPu, NAcC, and nucleus accumbens shell (NAcSh). nacc 135-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 16300424-4 2005 In a separate experiment, a low, behaviorally relevant dose of cocaine was found to increase c-Fos immunoreactivity in the medial CPu, NAcC, and nucleus accumbens shell (NAcSh). nacsh 170-175 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 16300424-5 2005 The selective 5-HT2A receptor antagonist M100907 significantly attenuated cocaine-induced c-Fos expression in the medial CPu and in the NAcSh. Cocaine 74-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 16300424-5 2005 The selective 5-HT2A receptor antagonist M100907 significantly attenuated cocaine-induced c-Fos expression in the medial CPu and in the NAcSh. nacsh 136-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 16140209-6 2005 Pretreatment of PC12 cells with c-fos antisense oligonucleotide abolished the NO-induced increase in DNA binding of AP-1 and upregulation of COX-2 expression. Oligonucleotides 48-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 16159376-6 2005 The number of double-labelled tyrosine hydroxylase/Fos immunoreactive neurones in locus coeruleus was significantly higher at 14.00 h of oestrus, suggesting an increase in its activity preceding the prolactin surge that generally occurs at 15.00 h. Therefore, the increase in locus coeruleus activity on the afternoon of oestrus supports the data obtained with bilateral lesion of this nucleus, suggesting a stimulatory role of locus coeruleus norepinephrine in the genesis of the secondary surge of prolactin. Norepinephrine 444-458 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 16002144-4 2005 BOTOX also dose dependently reduced the number of formalin-induced Fos-like immunoreactive cells in the dorsal horn of the spinal cord and significantly (15 and 30 U/kg) inhibited the excitation of wide dynamic range neurons of the dorsal horn in Phase II but not Phase I of the formalin response. Formaldehyde 50-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 16039738-0 2005 Enhanced cardiovascular alteration and Fos expression induced by central salt loading in a conscious rat transgenic for the metallothionein-vasopressin fusion gene. Salts 73-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 16039738-3 2005 Central HS (0.3, 0.67, or 1.0M NaCl, 1 microl/min for 20 min) significantly increased the mean arterial blood pressure (MABP) and Fos-like immunoreactivity (FLI) in the paraventricular nucleus (PVN) and supraoptic nucleus (SON) of the hypothalamus, the area postrema (AP), the median preoptic nucleus (MnPO), and the organum vasculosum laminae terminalis (OVLT) in both Tg and control rats. Sodium Chloride 31-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-133 16122826-4 2005 Hydroxylamine (an inhibitor of cystathionine b-synthase) up-regulated the expression of c-fos and down-regulated the expression of GABA(B)R2, but did not change the expression of GABA(B)R1. Hydroxylamine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 16037947-6 2005 In addition, immunolabeling for tyrosine hydroxylase and/or Fos showed that the grafts reinnervated the surrounding striatal tissue with dopaminergic terminals, and induced the expression of Fos in the striatal neurons of the reinnervated area after administration of amphetamine to the host rat. Amphetamine 268-279 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 191-194 16140174-0 2005 Cyclic estradiol replacement attenuates stress-induced c-Fos expression in the PVN of ovariectomized rats. cyclic estradiol 0-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 16002220-0 2005 Increased c-Fos expression in the medial part of the lateral habenula during cue-evoked heroin-seeking in rats. Heroin 88-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-15 16002220-8 2005 Findings showed that heroin-associated conditioned stimuli could induce robust heroin-seeking behavior that was associated with increased c-Fos immunoreactivity in the medial part of the LHb. Heroin 21-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 16002220-8 2005 Findings showed that heroin-associated conditioned stimuli could induce robust heroin-seeking behavior that was associated with increased c-Fos immunoreactivity in the medial part of the LHb. Heroin 79-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 16002220-8 2005 Findings showed that heroin-associated conditioned stimuli could induce robust heroin-seeking behavior that was associated with increased c-Fos immunoreactivity in the medial part of the LHb. 5-[[[(2~{r},3~{s},4~{r},5~{r})-5-(6-Aminopurin-9-Yl)-3,4-Bis(Oxidanyl)oxolan-2-Yl]methylamino]methyl]-4-Azanyl-1-(Methoxymethyl)pyrimidin-2-One 187-190 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 16098547-4 2005 Oleate also increased Fos-LI in the myenteric and submucosal plexuses of the duodenum and the jejunum, but not the ileum, while glucose only increased Fos-LI in the submucosal plexus of the ileum. Glucose 128-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-154 16098547-5 2005 The CCK(1) receptor antagonist, lorglumide, abolished Fos-LI in the DVC following infusions of either oleate or glucose. lorglumide 32-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 16098547-5 2005 The CCK(1) receptor antagonist, lorglumide, abolished Fos-LI in the DVC following infusions of either oleate or glucose. cyqualon 68-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 16098547-5 2005 The CCK(1) receptor antagonist, lorglumide, abolished Fos-LI in the DVC following infusions of either oleate or glucose. Oleic Acid 102-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 16098547-5 2005 The CCK(1) receptor antagonist, lorglumide, abolished Fos-LI in the DVC following infusions of either oleate or glucose. Glucose 112-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 16098547-6 2005 In addition, lorglumide attenuated oleate-induced Fos-LI in the myenteric and submucosal plexuses of the duodenum and jejunum. lorglumide 13-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 16098547-6 2005 In addition, lorglumide attenuated oleate-induced Fos-LI in the myenteric and submucosal plexuses of the duodenum and jejunum. Oleic Acid 35-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 16098547-8 2005 These data confirm previous reports indicating that CCK(1) receptors mediate increased DVC Fos-LI following intestinal infusion of oleate or glucose. Oleic Acid 131-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-94 16098547-8 2005 These data confirm previous reports indicating that CCK(1) receptors mediate increased DVC Fos-LI following intestinal infusion of oleate or glucose. Glucose 141-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-94 16098547-9 2005 CCK(1) receptors also contribute to increased Fos-LI in enteric neurons following intestinal oleate infusion. Oleic Acid 93-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 16153612-1 2005 Acute administration of the dopamine-depleting agent reserpine (10 mg/kg) induces Fos expression in striatopallidal neurons of intact rats-an effect that is blocked by pretreatment with the D2 agonist quinpirole (0.5 mg/kg). Dopamine 28-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 16153612-1 2005 Acute administration of the dopamine-depleting agent reserpine (10 mg/kg) induces Fos expression in striatopallidal neurons of intact rats-an effect that is blocked by pretreatment with the D2 agonist quinpirole (0.5 mg/kg). Reserpine 53-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 16153612-1 2005 Acute administration of the dopamine-depleting agent reserpine (10 mg/kg) induces Fos expression in striatopallidal neurons of intact rats-an effect that is blocked by pretreatment with the D2 agonist quinpirole (0.5 mg/kg). Quinpirole 201-211 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 16153612-2 2005 Systemic administration of the muscarinic antagonist scopolamine (50 mg/kg) partially attenuates reserpine-mediated striatal Fos expression. Scopolamine 53-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-128 16153612-2 2005 Systemic administration of the muscarinic antagonist scopolamine (50 mg/kg) partially attenuates reserpine-mediated striatal Fos expression. Reserpine 97-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-128 15994850-4 2005 Intraplantar administration of formalin mimicked inflammatory pain, and its effects were assessed using immunohistochemical (c-Fos staining) and behavioural paradigms. Formaldehyde 31-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 16162281-2 2005 We have previously found that gastric mucosal challenge with excess HCl is signalled to the rat brainstem, but not spinal cord, as visualized by expression of c-fos messenger ribonucleic acid (mRNA), a surrogate marker of neuronal excitation. Hydrochloric Acid 68-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-164 16162281-4 2005 RESULTS: Rats were treated intragastrically with vehicle, HCl or capsaicin, activation of neurons in the brainstem and spinal cord was visualized by in situ hybridization autoradiography for c-fos mRNA, and gastric emptying deduced from the retention of intragastrically administered fluid. Hydrochloric Acid 58-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 191-196 16162281-5 2005 Relative to vehicle, HCl (0.5 M) and capsaicin (3.2 mM) increased c-fos transcription in the nucleus tractus solitarii by factors of 7.0 and 2.1, respectively. Hydrochloric Acid 21-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 16162281-5 2005 Relative to vehicle, HCl (0.5 M) and capsaicin (3.2 mM) increased c-fos transcription in the nucleus tractus solitarii by factors of 7.0 and 2.1, respectively. Capsaicin 37-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 16162281-6 2005 Capsaicin also caused a 5.2-fold rise of c-fos mRNA expression in lamina I of the caudal thoracic spinal cord, although the number of c-fos mRNA-positive cells in this lamina was very small. Capsaicin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 16162281-6 2005 Capsaicin also caused a 5.2-fold rise of c-fos mRNA expression in lamina I of the caudal thoracic spinal cord, although the number of c-fos mRNA-positive cells in this lamina was very small. Capsaicin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-139 15879055-8 2005 Interestingly, the AT(2) receptor antagonist PD-123319 alone slightly enhanced the expression of c-Fos, c-Jun, and Krox-24 in different populations of neurons of the paraventricular nucleus. PD 123319 45-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 16112401-3 2005 Forty-micrometer brainstem sections containing the area postrema, nucleus of the solitary tract, and the dorsal motor nucleus of the vagus, and myenteric neurons of the duodenum, jejunum, and ileum underwent a diaminobenzidine reaction enhanced with nickel to reveal Fos-LI. 4,4'-Dihydrazino-biphenyl 210-226 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 267-270 15979947-4 2005 In Long-Evans Tokushima Otsuka (LETO) rats (controls), c-fos mRNA was detected abundantly in those nuclei 30 min after iv administration of CCK-8 (20 microg/kg). cholecystokinin 8 140-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 15890444-5 2005 Using an immediate early gene product Fos as a reporter of inducible gene expression, cyclosporin A was found to upregulate Fos expression in the dorsal striatum. Cyclosporine 86-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 15890444-5 2005 Using an immediate early gene product Fos as a reporter of inducible gene expression, cyclosporin A was found to upregulate Fos expression in the dorsal striatum. Cyclosporine 86-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-127 15979572-3 2005 In cultured rat cortical neurons, over-expression of truncated dentatorubral-pallidoluysian atrophy proteins containing expanded polyQ, which form aggregation bodies in nucleus, reduced the calcium (Ca(2+)) signal-mediated transcriptional activation of brain-derived neurotrophic factor, c-fos, and pituitary adenylate cyclase-activating polypeptide gene promoters in a dose-dependent manner. Calcium 190-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 288-293 15973680-0 2005 Hippocampal granule cell activity and c-Fos expression during spontaneous seizures in awake, chronically epileptic, pilocarpine-treated rats: implications for hippocampal epileptogenesis. Pilocarpine 116-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 15955413-0 2005 Cocaine pre-exposure enhances CRF-induced expression of c-fos mRNA in the central nucleus of the amygdala: an effect that parallels the effects of cocaine pre-exposure on CRF-induced locomotor activity. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 15955413-0 2005 Cocaine pre-exposure enhances CRF-induced expression of c-fos mRNA in the central nucleus of the amygdala: an effect that parallels the effects of cocaine pre-exposure on CRF-induced locomotor activity. Cocaine 147-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 15955417-5 2005 Pretreatment with prazosin (5 mg/kg) effectively suppressed the Fos expression induced by LPS (5 microg/kg), whereas L-NAME (30 mg/kg) did not influence the Fos expression in the AVP neurons induced by LPS (0.25, 0.5, 1, 5 microg/kg). Prazosin 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 16038632-0 2005 Puerarin reduces increased c-fos, c-jun, and type IV collagen expression caused by high glucose in glomerular mesangial cells. puerarin 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 16038632-4 2005 The aim of this study is to investigate the effect of puerarin on c-fos, c-jun and CoIV expression in GMC cultured in medium containing 5.6 or 27.8 mmol/L glucose. puerarin 54-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 16038632-8 2005 RESULTS: Puerarin (10(-5) mmol/L) significantly ameliorated the high-glucose effect on c-fos, c-jun and CoIV expression. puerarin 9-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 16038632-8 2005 RESULTS: Puerarin (10(-5) mmol/L) significantly ameliorated the high-glucose effect on c-fos, c-jun and CoIV expression. Glucose 69-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 16038632-10 2005 CONCLUSION: Our results suggest that reduced PKC activity and expression of c-fos and c-jun in GMC might participate in the mechanisms underlying the therapeutic effect of puerarin on diabetic nephropathy. puerarin 172-180 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 15921658-6 2005 In contrast, unilateral injections of muscimol into the AcbSh consistently increased Fos expression in several brain regions. Muscimol 38-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 15905220-4 2005 To test this hypothesis, we quantified expression of Fos-like immunoreactivity (Fos) in the dorsal vagal complex (DVC) of capsaicin-treated (Cap) and control rats (Veh), following gastric balloon distension alone and in combination with CCK injection. Capsaicin 122-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-78 15905220-4 2005 To test this hypothesis, we quantified expression of Fos-like immunoreactivity (Fos) in the dorsal vagal complex (DVC) of capsaicin-treated (Cap) and control rats (Veh), following gastric balloon distension alone and in combination with CCK injection. Capsaicin 122-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 15905220-8 2005 Furthermore, CCK"s enhancement of distension-induced Fos in Cap rats was reversed by the selective CCK-A receptor antagonist lorglumide. lorglumide 125-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 16179556-3 2005 In the present study, we evaluated the effect of increasing doses of cocaine on the expression of immediate early genes (IEGs), c-fos and c-jun, and closely related transcription factors, SP-1 and NF-kbeta, at 24 h after the exposure to cocaine (50, 100, 200, 500, 1000, 2500 microM) in NGF-differentiated PC12 cells. Cocaine 69-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-133 16179556-4 2005 Cocaine (50-500 microM) resulted in significant induction of the expression of c-fos, c-jun, SP-1, and NF-kbeta. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 16044040-3 2005 Pretreatment with morphine 30 min before pulp exposure dose-dependently (2.5, 5, and 10 mg/kg subcutaneously) reduced Fos expression in subnucleus caudalis whereas pretreatment with ibuprofen (10-100 mg/kg subcutaneously) did not significantly affect Fos expression. Ibuprofen 182-191 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 16044040-3 2005 Pretreatment with morphine 30 min before pulp exposure dose-dependently (2.5, 5, and 10 mg/kg subcutaneously) reduced Fos expression in subnucleus caudalis whereas pretreatment with ibuprofen (10-100 mg/kg subcutaneously) did not significantly affect Fos expression. Ibuprofen 182-191 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 251-254 15993445-4 2005 Pre-treatment with naloxone methiodide decreased (15%) IL-1beta-induced Fos-immunoreactivity (Fos-IR) in medial parvocellular paraventricular nucleus (mPVN) corticotropin-releasing hormone (CRH) neurons but increased responses in the ventrolateral medulla (VLM) C1 (65%) and nucleus tractus solitarius (NTS) A2 (110%) catecholamine cell groups and area postrema (136%). N-methylnaloxone 19-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 15993445-4 2005 Pre-treatment with naloxone methiodide decreased (15%) IL-1beta-induced Fos-immunoreactivity (Fos-IR) in medial parvocellular paraventricular nucleus (mPVN) corticotropin-releasing hormone (CRH) neurons but increased responses in the ventrolateral medulla (VLM) C1 (65%) and nucleus tractus solitarius (NTS) A2 (110%) catecholamine cell groups and area postrema (136%). N-methylnaloxone 19-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 15993445-4 2005 Pre-treatment with naloxone methiodide decreased (15%) IL-1beta-induced Fos-immunoreactivity (Fos-IR) in medial parvocellular paraventricular nucleus (mPVN) corticotropin-releasing hormone (CRH) neurons but increased responses in the ventrolateral medulla (VLM) C1 (65%) and nucleus tractus solitarius (NTS) A2 (110%) catecholamine cell groups and area postrema (136%). Catecholamines 318-331 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 15993445-6 2005 We next determined the effect of blocking both central and peripheral opioid receptors with naloxone and, when compared to the naloxone methiodide pre-treated group, a further 60% decrease in Fos-IR mPVN CRH neurons induced by IL-1beta was detected, which was attributed to block of central opioid receptors. N-methylnaloxone 127-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 192-195 15993445-7 2005 Similar comparisons also detected decreases in Fos-IR neurons induced by IL-1beta in the VLM A1, VLM C1 and NTS A2 catecholamine cell groups, area postrema, and parabrachial nucleus. Catecholamines 115-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 15993445-8 2005 In contrast, pre-treatment with naloxone increased Fos-IR neurons in CeA (98%) and dorsal BNST (72%). Naloxone 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 15982643-2 2005 Pretreatment with the antimuscarinic drugs scopolamine and atropine was able to greatly suppress novelty-induced Fos expression at these sites. Scopolamine 43-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-116 15982643-2 2005 Pretreatment with the antimuscarinic drugs scopolamine and atropine was able to greatly suppress novelty-induced Fos expression at these sites. Atropine 59-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-116 15973150-4 2005 In rats killed at the end of a 90 min ejection of bicuculline, Fos expression was induced in approximately 28% of the neurons immunoreactive for hypocretin and in approximately 3% of the neurons immunostained for melanin-concentrating hormone within the ejection site. Bicuculline 50-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 15960884-12 2005 Intrathecal injection of U0126 or ERK antisense ODN markedly suppressed the increase of CCI-induced pERK, pCREB and c-Fos expression in the spinal cord. U 0126 25-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 15960884-12 2005 Intrathecal injection of U0126 or ERK antisense ODN markedly suppressed the increase of CCI-induced pERK, pCREB and c-Fos expression in the spinal cord. CCI 88-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 15946158-5 2005 The data indicate that, 2 h after 5TG treatment, Fos immunoexpression by rostral preoptic GnRH neurones and plasma LH levels were diminished relative to the vehicle-treated controls, and that inhibitory effects of 5TG on these parameters were attenuated by pretreatment with bicuculline, but not phaclofen. Bicuculline 275-286 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 15946158-5 2005 The data indicate that, 2 h after 5TG treatment, Fos immunoexpression by rostral preoptic GnRH neurones and plasma LH levels were diminished relative to the vehicle-treated controls, and that inhibitory effects of 5TG on these parameters were attenuated by pretreatment with bicuculline, but not phaclofen. phaclofen 296-305 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 15988009-6 2005 The basal protein and mRNA levels and nuclear binding activities of c-Jun, c-Fos, p50, and p65 were lower in F1 and F2 cells and exhibited a blunted response to TBH. 2-tert-butylhydroquinone 161-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 16342423-4 2005 The downstream effects of blocking PKC alpha activity by exposure to Go6976 include inhibition of IGF1-stimuated PI3 kinase activity and reduced IGF1-stimulated c-fos expression in the adult cardiomyocytes. Go 6976 69-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 15714225-0 2005 Stress-induced c-Fos expression is differentially modulated by dexamethasone, diazepam and imipramine. Dexamethasone 63-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 15714225-0 2005 Stress-induced c-Fos expression is differentially modulated by dexamethasone, diazepam and imipramine. Diazepam 78-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 15714225-0 2005 Stress-induced c-Fos expression is differentially modulated by dexamethasone, diazepam and imipramine. Imipramine 91-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 15714225-10 2005 Dexamethasone reduced stress-induced c-Fos expression in SS cortex, hippocampus, paraventricular nucleus of the hypothalamus (PVH), and locus coeruleus (LC), whereas diazepam reduced c-Fos staining in the SS cortex, hippocampus, bed nucleus of stria terminalis, septal area, and hypothalamus (preoptic area and supramammillary nucleus). Dexamethasone 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 15714225-12 2005 We conclude that dexamethasone, diazepam and imipramine differentially modulate stress-induced Fos expression. Dexamethasone 17-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 15714225-12 2005 We conclude that dexamethasone, diazepam and imipramine differentially modulate stress-induced Fos expression. Diazepam 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 15714225-12 2005 We conclude that dexamethasone, diazepam and imipramine differentially modulate stress-induced Fos expression. Imipramine 45-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 16041643-4 2005 Kaempferol also inhibited the c-fos mRNA expression induced by PDGF-BB concentration-dependently. kaempferol 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 15858832-2 2005 We have found that repeated stimulation of the rat prelimbic cortex with picrotoxin (0.25 microg/0.25 microl, five injections on alternate days followed by 7-day withdrawal) contributed to increase c-Fos protein expression in the striosomes of the dorsolateral striatum, while producing the opposite effect in the matrix compartment, after a single exposure to cocaine (25 mg/kg). Picrotoxin 73-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 198-203 16078581-8 2005 FOS positive neurons were induced by hypoxia and mainly existed in the nucleus of solitary tract, area postrema, hypoglossal nucleus, lateral reticular nucleus, inferior olivary nucleus, nucleus raphe pallidus, facial nucleus, trapezoid nucleus, but in the group of hypoxia plus TMP, the level of FOS immunoreactivity decreased remarkably, compared with the group of hypoxia (P<0.05). tetramethylpyrazine 279-282 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 16078581-9 2005 CONCLUSION: TMP has protective effect on the respiratory inhibition induced by hypoxia and FOS may be involved in the protection. tetramethylpyrazine 12-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-94 15905220-1 2005 Capsaicin treatment destroys vagal afferent C fibers and markedly attenuates reduction of food intake and induction of hindbrain Fos expression by CCK. Capsaicin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-132 16173470-10 2005 All dosages of L365,260 failed to attenuate this increase; however, injection of devazepide attenuated the increase in Fos-LI. Devazepide 81-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-122 16044040-0 2005 Effects of pre-emptive morphine, ibuprofen or local anesthetic on fos expression in the spinal trigeminal nucleus following tooth pulp exposure in the rat. Morphine 23-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 16044040-0 2005 Effects of pre-emptive morphine, ibuprofen or local anesthetic on fos expression in the spinal trigeminal nucleus following tooth pulp exposure in the rat. Ibuprofen 33-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 16044040-3 2005 Pretreatment with morphine 30 min before pulp exposure dose-dependently (2.5, 5, and 10 mg/kg subcutaneously) reduced Fos expression in subnucleus caudalis whereas pretreatment with ibuprofen (10-100 mg/kg subcutaneously) did not significantly affect Fos expression. Morphine 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 16044040-3 2005 Pretreatment with morphine 30 min before pulp exposure dose-dependently (2.5, 5, and 10 mg/kg subcutaneously) reduced Fos expression in subnucleus caudalis whereas pretreatment with ibuprofen (10-100 mg/kg subcutaneously) did not significantly affect Fos expression. Morphine 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 251-254 16184402-4 2005 We previously reported that ET-1 induces ROS generation via the ET(A) receptor and ROS modulates c-fos gene expression. ros 83-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 16184402-7 2005 Trilinolein (1 and 10 microM) inhibited ET-1-induced c-fos gene expression in cardiomyocytes. trilinolein 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 16025001-4 2005 RESULTS: The number of Fos positive cells significantly increased in the paraventricular nucleus of the hypothalamus (PVN, 191 +/- 63 versus 66 +/- 18), pontine locus coeruleus (LC, 53 +/- 19 versus 5 +/- 2), brain medullary dorsal motor nucleus of the vagus (DMV, 26 +/- 4 versus 1 +/- 0), and nucleus tractus solitarii (NTS, 38 +/- 3 versus 10 +/- 35) in rats with hyperinsulinemic/hypoglycemic clamp compared with the controls. (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 260-263 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 16025001-5 2005 Maintaining blood glucose levels within physiological range by hyperinsulinemic/euglycemic clamp prevented insulin infusion-induced Fos expression in the PVN, DMV, and NTS. Glucose 18-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-135 16025001-5 2005 Maintaining blood glucose levels within physiological range by hyperinsulinemic/euglycemic clamp prevented insulin infusion-induced Fos expression in the PVN, DMV, and NTS. (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 159-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-135 16268501-6 2005 Compared with MNU-treated rats, the gene expression of c-jun and c-fos was time-dependently down-regulated in ligustrazine-treated group. Methylnitrosourea 14-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 16268501-6 2005 Compared with MNU-treated rats, the gene expression of c-jun and c-fos was time-dependently down-regulated in ligustrazine-treated group. tetramethylpyrazine 110-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 16268501-7 2005 CONCLUSION: Ligustrazine injection partially protects against MNU-induced retinal damage by down-modulating the expression of c-jun and c-fos genes to inhibit apoptosis of photoreceptor cells. tetramethylpyrazine 12-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 16268501-7 2005 CONCLUSION: Ligustrazine injection partially protects against MNU-induced retinal damage by down-modulating the expression of c-jun and c-fos genes to inhibit apoptosis of photoreceptor cells. Methylnitrosourea 62-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 16051541-0 2005 c-Fos antisense oligodeoxynucleotide offsets behavioral nociceptive responses and both up-regulations of c-Fos protein and dynorphin a (1-8) in dorsal horn: a study using the formalin test in rats. Oligodeoxyribonucleotides 16-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 16051541-0 2005 c-Fos antisense oligodeoxynucleotide offsets behavioral nociceptive responses and both up-regulations of c-Fos protein and dynorphin a (1-8) in dorsal horn: a study using the formalin test in rats. Oligodeoxyribonucleotides 16-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 16051541-0 2005 c-Fos antisense oligodeoxynucleotide offsets behavioral nociceptive responses and both up-regulations of c-Fos protein and dynorphin a (1-8) in dorsal horn: a study using the formalin test in rats. Formaldehyde 175-183 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 15693018-8 2005 The increase in PTH stimulation of c-jun, c-fos, and MMP-13 in G(11)alpha-transfected cells were all blocked by bisindolylmaleimide I, a selective inhibitor of PKC. bisindolylmaleimide I 112-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 15921658-8 2005 Fos synthesis in the LH was increased only on the side of the brain ipsilateral to the muscimol injection. Muscimol 87-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 15829701-9 2005 In SOCS-3 knockdown studies, antisense oligonucleotides inhibited the expression of SOCS-3 and increased the Ang II-induced STAT activation and c-Fos/c-Jun expression, then resulting in a more severe renal damage. Oligonucleotides 39-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-149 15946158-5 2005 The data indicate that, 2 h after 5TG treatment, Fos immunoexpression by rostral preoptic GnRH neurones and plasma LH levels were diminished relative to the vehicle-treated controls, and that inhibitory effects of 5TG on these parameters were attenuated by pretreatment with bicuculline, but not phaclofen. 5-thio-D-glucose 34-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 15837002-0 2005 Immunohistochemical localization of toluene-induced c-Fos protein expression in the rat brain. Toluene 36-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 15837002-4 2005 Quantitative analysis of Fos-immunoreactive neurons indicated toluene dose-related induced c-Fos immunoreactivity in the majority of structures examined. Toluene 62-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 15837002-4 2005 Quantitative analysis of Fos-immunoreactive neurons indicated toluene dose-related induced c-Fos immunoreactivity in the majority of structures examined. Toluene 62-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 15837002-7 2005 The data demonstrate that toluene dose-related induced a unique pattern of c-Fos immunoreactivity. Toluene 26-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 15837002-8 2005 The widespread distribution of toluene-induced c-Fos expression seen in this study can be linked to the profound alterations in physiological function and behavior produced by this solvent. Toluene 31-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 15927549-7 2005 Pretreatment with SR 141716A inhibited the WIN 55,212-2 induced c-Fos expression, while the antagonist alone did not affect c-Fos expression. Rimonabant 18-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 15862908-8 2005 The data clearly showed that PrRP cells in DES-administered rats significantly suppressed c-Fos accumulation induced by stress. Diethylstilbestrol 43-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 15976532-8 2005 Brain c-Fos expression was increased in hippocampus, prefrontal cortex and visual cortex in rats trained on the DMP compared to the RAN task. dmp 112-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 6-11 15976532-9 2005 Furthermore, in the DMP-trained rats, hippocampal c-Fos expression was lower in ethanol-exposed rats. dmp 20-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 15976532-9 2005 Furthermore, in the DMP-trained rats, hippocampal c-Fos expression was lower in ethanol-exposed rats. Ethanol 80-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 15976532-10 2005 CONCLUSIONS: These results suggest that the short-term memory impairment of ethanol-exposed rats 1) can be improved slightly by an increase in encoding time and 2) is related to a decrease in c-Fos expression in the hippocampus. Ethanol 76-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 192-197 15978010-3 2005 We examined the ability of dopamine agonists to activate orexin neurons in the rat, as reflected by induction of Fos. Dopamine 27-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-116 15978010-4 2005 The mixed dopamine agonist apomorphine increased Fos expression in orexin cells, with a greater effect on orexin neurons located medial to the fornix. Dopamine 10-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 15978010-4 2005 The mixed dopamine agonist apomorphine increased Fos expression in orexin cells, with a greater effect on orexin neurons located medial to the fornix. Apomorphine 27-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 15978010-5 2005 Both the selective D1-like agonist, A-77636, and the D2-like agonist, quinpirole, also induced Fos in orexin cells, suggesting that stimulation of either receptor subtype is sufficient to activate orexin neurons. A 77636 36-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 15978010-5 2005 Both the selective D1-like agonist, A-77636, and the D2-like agonist, quinpirole, also induced Fos in orexin cells, suggesting that stimulation of either receptor subtype is sufficient to activate orexin neurons. Quinpirole 70-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 15916815-0 2005 Methylphenidate differentially regulates c-fos and fosB expression in the developing rat striatum. Methylphenidate 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 15896499-5 2005 Here, we report the alterations in mRNAs levels of c-Fos, JunB, and Krox20 proteins induced in the rat brain and liver by the acute exposure to 3-NPA at 30 mg/kg, s.c. 3-nitropropionic acid 144-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 15904713-5 2005 The activity in the nucleus of the solitary tract (NTS), a brainstem relay mediating inhibition of intake, judged by the expression of c-fos-like immunoreactivity, was significantly increased after treatment with CCK-8 or NPY to approximately the same extent. Sincalide 213-218 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 15904717-3 2005 Expression of c-Fos in the amygdala (the central nucleus, CeA; the medial nucleus, MeA; the basolateral nucleus, BLA) following naloxone-precipitated withdrawal and the CPA test was examined using a range of naloxone doses (0.02, 0.05, 0.1, 0.2, 0.5 and 1.0 mg/kg). mea 83-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 15904717-3 2005 Expression of c-Fos in the amygdala (the central nucleus, CeA; the medial nucleus, MeA; the basolateral nucleus, BLA) following naloxone-precipitated withdrawal and the CPA test was examined using a range of naloxone doses (0.02, 0.05, 0.1, 0.2, 0.5 and 1.0 mg/kg). Naloxone 128-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 15904717-5 2005 In CeA, but not MeA with high-level constitutive neuronal activity, the naloxone-induced modification in c-Fos immunoreactivity following morphine pretreatment exhibited a dose-dependent pattern similar to that seen in the behavioral study. Naloxone 72-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 15904717-5 2005 In CeA, but not MeA with high-level constitutive neuronal activity, the naloxone-induced modification in c-Fos immunoreactivity following morphine pretreatment exhibited a dose-dependent pattern similar to that seen in the behavioral study. Morphine 138-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 15846122-9 2005 Rapid (by 3 h) and transient increases in c-fos and jun-B mRNA levels were observed after E2 treatment; however, c-fos and jun-B induction by 4OH was highly dose dependent, and higher 4OH doses induced rapid but persistent proto-oncogene expression in vivo. Estetrol 142-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 15722198-4 2005 The c-Fos induction was greatly diminished by Go6976 or PD098059, and completely abolished when combined. Go 6976 46-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 15722198-4 2005 The c-Fos induction was greatly diminished by Go6976 or PD098059, and completely abolished when combined. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 56-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 15929744-6 2005 Refeeding with sugar alone for 2 h at the end of a 48-h fast also reduced the potentiated Fos response in fasting, indicating that elevated blood glucose can influence the central responsiveness to ghrelin/GHS. Sugars 15-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 15929744-6 2005 Refeeding with sugar alone for 2 h at the end of a 48-h fast also reduced the potentiated Fos response in fasting, indicating that elevated blood glucose can influence the central responsiveness to ghrelin/GHS. Glucose 146-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 15935212-10 2005 In summary, iodine deficiency and hypothyroidism during critical periods of brain development impair LTP induction and decrease the expression of c-fos and c-jun proteins in hippocampus. Iodine 12-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-151 16044914-0 2005 [Intrathecal injection of muscarinic receptors or GDNF antisense oligonucleotides inhibits the increase of c-Fos expression in locus coeruleus of morphine-withdrawal rats]. Oligonucleotides 65-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 16044914-0 2005 [Intrathecal injection of muscarinic receptors or GDNF antisense oligonucleotides inhibits the increase of c-Fos expression in locus coeruleus of morphine-withdrawal rats]. Morphine 146-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 15880490-9 2005 By using basal c-Fos expression as a marker for cellular activation we found a significant reduction in c-Fos expression in the central nucleus of the inferior colliculus in iron-adequate rat pups exposed to CO. By contrast, rather than being reduced, c-Fos expression in the ARIDCO group is the same as for controls. Iron 174-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 15880490-9 2005 By using basal c-Fos expression as a marker for cellular activation we found a significant reduction in c-Fos expression in the central nucleus of the inferior colliculus in iron-adequate rat pups exposed to CO. By contrast, rather than being reduced, c-Fos expression in the ARIDCO group is the same as for controls. Iron 174-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 15880490-9 2005 By using basal c-Fos expression as a marker for cellular activation we found a significant reduction in c-Fos expression in the central nucleus of the inferior colliculus in iron-adequate rat pups exposed to CO. By contrast, rather than being reduced, c-Fos expression in the ARIDCO group is the same as for controls. Iron 174-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 15985704-1 2005 In the present study we wanted to check whether the expression of the c-Fos protein (the marker of cellular activity) appears in cells containing calcium-binding proteins (CaBPs) in animals exposed to the open field test. Calcium 146-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 15985704-9 2005 It seems that cells containing CaBPs are not directly involved in the response to aversive stimuli but cells containing those calcium-binding proteins might influence directly c-Fos positive neurons of PC. Calcium 126-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 176-181 15910767-14 2005 injection of either 10 microg of NPW23 or 10 microg of NPB significantly suppressed the expression of Fos-like immunoreactivity of the L4-5 spinal dorsal horn induced by paw formalin injection. Formaldehyde 174-182 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 15854742-7 2005 injection of losartan, angiotensin II type 1 (AT1) receptor antagonist (5 microg/microl), the excitatory effect of peripheral hypertonic stimulation on PVN neurons with phasic activity was inhibited significantly, and the number of the neurons co-expressing Fos and AVP in PVN decreased significantly (P<0.001) as well. Losartan 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 258-261 15866553-0 2005 Sensitized attentional performance and Fos-immunoreactive cholinergic neurons in the basal forebrain of amphetamine-pretreated rats. Amphetamine 104-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 15866553-8 2005 In AMPH-pretreated and -challenged animals, an increased number of Fos-IR neurons was observed in the basal forebrain. Amphetamine 3-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 16044914-3 2005 The expression of c-Fos positive neurons in the LC increased in morphine-dependent rats and increased to a greater extent after the injection of naloxone (4mg/kg, ip) in morphine dependent rats. Morphine 64-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 16044914-3 2005 The expression of c-Fos positive neurons in the LC increased in morphine-dependent rats and increased to a greater extent after the injection of naloxone (4mg/kg, ip) in morphine dependent rats. Naloxone 145-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 15910798-1 2005 We studied the effects of the high-efficacy 5-hydroxytryptamine1A (5-HT1A) receptor agonist, F 13640 on both formalin-induced spinal cord c-Fos protein expression and pain behaviours in the rat. Formaldehyde 109-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 16044914-3 2005 The expression of c-Fos positive neurons in the LC increased in morphine-dependent rats and increased to a greater extent after the injection of naloxone (4mg/kg, ip) in morphine dependent rats. Morphine 170-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 16044914-4 2005 Intrathecal injection of M2AS-oligo or GDNFAS-oligo inhibited the increase of c-Fos expression in LC during morphine withdrawal, but there was no effect in case of M1AS-oligo. Morphine 108-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 15910798-4 2005 In the same animals, and in spite of the agent as in earlier data increasing the number of c-Fos labelled nuclei when it was administered alone, F 13640 markedly reduced the number of formalin-induced c-Fos labelled nuclei. Formaldehyde 184-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 201-206 15910798-6 2005 Co-operation occurred so that after the co-administration of F 13640 and formalin, c-Fos expression was inferior to that induced when either stimulation was administered alone. Formaldehyde 73-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 15926928-3 2005 Whereas c-Fos expression was increased in Orx neurons after SD, it was increased in MCH neurons after SR. We reasoned that Orx and MCH neurons could be differently modulated by noradrenaline (NA) and accordingly bear different adrenergic receptors (ARs). Norepinephrine 177-190 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-13 15872129-8 2005 We also examined the effect of systemic ketamine on the c-fos immunoreactivity in the spinal cord. Ketamine 40-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 15793830-6 2005 The atelocollagen and silicone tube groups demonstrated fewer c-fos-expressed cells in the spinal cord than the controls. Silicones 22-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 15857691-0 2005 Valproic acid, but not lamotrigine, suppresses seizure-induced c-fos and c-Jun mRNA expression. Valproic Acid 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 15785859-1 2005 We previously reported that nicotine withdrawal up-regulates transcription of some immediately early genes (IEGs), c-fos (Ichino et al., 1999) and egr1, nur77 (Ichino et al., 2002) in cultures of pheochromocytoma PC12 cells, which are of neuronal lineage. Nicotine 28-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 15857691-3 2005 We found that valproic acid (VPA), but not lamotrigine (LTG), was capable of suppressing seizure-induced c-fos and c-Jun mRNA expression in rats despite a similar anticonvulsant effect. Valproic Acid 14-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 15857691-3 2005 We found that valproic acid (VPA), but not lamotrigine (LTG), was capable of suppressing seizure-induced c-fos and c-Jun mRNA expression in rats despite a similar anticonvulsant effect. Valproic Acid 29-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 15830097-10 2005 Correlating with behavior results, the injection also markedly suppressed the increase of CCI-induced pCREB and c-Fos expression. CCI 90-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 15830100-9 2005 The results showed that intrathecal administration of CHE decreased the scores of morphine withdrawal, attenuated morphine withdrawal-induced allodynia and also inhibited the increase of Fos protein expression in the spinal cord of morphine withdrawal rats. Chlorides 54-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 187-190 15862622-0 2005 Methylphenidate regulates c-fos and fosB expression in multiple regions of the immature rat brain. Methylphenidate 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 15829640-9 2005 The CS induced Fos expression in CeA, and a majority of these Fos-positive neurons were also FG positive, indicating activation of the CeA-SNc pathway by the CS. Cesium 158-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 15637641-0 2005 Topography of methylphenidate (ritalin)-induced gene regulation in the striatum: differential effects on c-fos, substance P and opioid peptides. Methylphenidate 31-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 15637641-6 2005 of methylphenidate produced robust increases in the expression of the transcription factor c-fos and the neuropeptide substance P. Borderline effects were found with 2 mg/kg, but not with 0.5 mg/kg. Methylphenidate 3-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 15637641-11 2005 These results demonstrate that acute methylphenidate alters the expression of c-fos and substance P preferentially in the sensorimotor striatum. Methylphenidate 37-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 15804434-0 2005 Role of protein kinase C beta in phorbol ester-induced c-fos gene expression in neurons of normotensive and spontaneously hypertensive rat brains. Phorbol Esters 33-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 15804434-4 2005 PMA increased c-fos gene expression in neuronal cultures of Wistar rat brain and the PMA-induced c-fos gene expression was inhibited by the PKC inhibitor 1-(5-isoquinolinesulfonyl)-2-methylpiperazine dihydrochloride (H-7). Tetradecanoylphorbol Acetate 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 15804434-4 2005 PMA increased c-fos gene expression in neuronal cultures of Wistar rat brain and the PMA-induced c-fos gene expression was inhibited by the PKC inhibitor 1-(5-isoquinolinesulfonyl)-2-methylpiperazine dihydrochloride (H-7). Tetradecanoylphorbol Acetate 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 15804434-4 2005 PMA increased c-fos gene expression in neuronal cultures of Wistar rat brain and the PMA-induced c-fos gene expression was inhibited by the PKC inhibitor 1-(5-isoquinolinesulfonyl)-2-methylpiperazine dihydrochloride (H-7). H-7 dihydrochloride 154-215 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 15804434-4 2005 PMA increased c-fos gene expression in neuronal cultures of Wistar rat brain and the PMA-induced c-fos gene expression was inhibited by the PKC inhibitor 1-(5-isoquinolinesulfonyl)-2-methylpiperazine dihydrochloride (H-7). H-7 dihydrochloride 154-215 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 15804434-6 2005 In addition, the PMA-induced c-fos gene expression was inhibited by PKCbetaantisense oligonucleotides (AON) but not by PKCalpha and PKCgammaAONs. Oligonucleotides 85-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 15804434-7 2005 In SHR brain neuronal cultures, the PMA-induced c-fos gene expression was enhanced as compared with that of Wistar Kyoto rats (WKY), while basal c-fos gene expression was almost the same in both neuronal cultures. Tetradecanoylphorbol Acetate 36-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 15689199-5 2005 GTN infusion (2 microg/kg/min for 30 min) did not lead to enhanced c-fos LI after 2 h and 4 h, whereas capsaicin infusion caused a time- and dose-dependent expression of c-fos LI within laminae I and II of the TNC. Capsaicin 103-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-175 15829640-7 2005 After a test with the CS alone, the brains were prepared to assess FG labeling and CS-induced Fos expression in CeA with immunohistochemical procedures. Cesium 83-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 15829640-9 2005 The CS induced Fos expression in CeA, and a majority of these Fos-positive neurons were also FG positive, indicating activation of the CeA-SNc pathway by the CS. Cesium 4-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-18 15829640-9 2005 The CS induced Fos expression in CeA, and a majority of these Fos-positive neurons were also FG positive, indicating activation of the CeA-SNc pathway by the CS. Cesium 4-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 15659565-0 2005 Neuroanatomical targets of the organophosphate chlorpyrifos by c-fos immunolabeling. Organophosphates 31-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 15659565-0 2005 Neuroanatomical targets of the organophosphate chlorpyrifos by c-fos immunolabeling. Chlorpyrifos 47-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 15576664-0 2005 Subfornical organ disconnection alters Fos expression in the lamina terminalis, supraoptic nucleus, and area postrema after intragastric hypertonic NaCl. Sodium Chloride 148-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 15869485-4 2005 Fos expression in barosensitive neurones was stimulated by an intravenous infusion of the hypotensive agent sodium nitroprusside. Nitroprusside 108-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 15842237-3 2005 Although some studies report that steroid hormones regulate the expression of the inducible transcription factor, Fos, in developing brain, it is not known if there is a sex difference in Fos expression. Steroids 34-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 15700284-3 2005 Priming induction using three injections with D1/D2 agonist apomorphine (0.5 mg/kg) or D1 agonist SKF38393 (10 mg/kg) allows priming expression, robust contralateral rotational behavior and striatal Fos expression, following a challenge with the D2 agonist quinpirole (0.25 mg/kg). Apomorphine 60-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-202 15700284-3 2005 Priming induction using three injections with D1/D2 agonist apomorphine (0.5 mg/kg) or D1 agonist SKF38393 (10 mg/kg) allows priming expression, robust contralateral rotational behavior and striatal Fos expression, following a challenge with the D2 agonist quinpirole (0.25 mg/kg). 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 98-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-202 15700284-6 2005 In contrast, administration of the AMPA antagonist 2,3-dihydroxy-6-nitro-7sulfamoyl-benzo[f]quinoxaline (NBQX) (5 or 10 mg/kg) potentiated apomorphine- and SKF38393-priming of quinpirole-mediated striatal Fos expression, but had no effect on their priming of quinpirole-mediated rotational behavior. 2,3-dioxo-6-nitro-7-sulfamoylbenzo(f)quinoxaline 51-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 205-208 15700284-6 2005 In contrast, administration of the AMPA antagonist 2,3-dihydroxy-6-nitro-7sulfamoyl-benzo[f]quinoxaline (NBQX) (5 or 10 mg/kg) potentiated apomorphine- and SKF38393-priming of quinpirole-mediated striatal Fos expression, but had no effect on their priming of quinpirole-mediated rotational behavior. 2,3-dioxo-6-nitro-7-sulfamoylbenzo(f)quinoxaline 105-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 205-208 15698898-4 2005 We found that nearly 35% of the histamine neurons from the tuberomammillary nucleus were Fos-immunoreactive immediately before mealtime. Histamine 32-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 15758184-7 2005 Furthermore, ERK1/2, when only coactivated by both IP3/Ca2+- and Homer1b/c-dependent pathways, showed the ability to phosphorylate two transcription factors, Elk-1 and cAMP response element-binding protein, and thereby facilitated c-Fos expression. Cyclic AMP 168-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 231-236 15692058-7 2005 SSP-saporin treatment also decreased c-fos expression in the dorsal horn of the spinal cord induced by instillation of capsaicin into the bladder. ssp-saporin 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 15692058-7 2005 SSP-saporin treatment also decreased c-fos expression in the dorsal horn of the spinal cord induced by instillation of capsaicin into the bladder. Capsaicin 119-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 15689199-6 2005 Surprisingly, GTN attenuated capsaicin-induced c-fos expression by 64%. Nitroglycerin 14-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 15689199-6 2005 Surprisingly, GTN attenuated capsaicin-induced c-fos expression by 64%. Capsaicin 29-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 15689199-7 2005 The nitric oxide synthase (NOS) inhibitor L-NAME (5 and 50 mg/kg) reduced capsaicin-induced c-fos LI dose dependently (reduction by 13% and 59%). NG-Nitroarginine Methyl Ester 42-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 15689199-7 2005 The nitric oxide synthase (NOS) inhibitor L-NAME (5 and 50 mg/kg) reduced capsaicin-induced c-fos LI dose dependently (reduction by 13% and 59%). Capsaicin 74-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 15689199-10 2005 Both GTN and L-NAME reduced capsaicin-induced c-fos LI. Nitroglycerin 5-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 15689199-10 2005 Both GTN and L-NAME reduced capsaicin-induced c-fos LI. NG-Nitroarginine Methyl Ester 13-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 15689199-10 2005 Both GTN and L-NAME reduced capsaicin-induced c-fos LI. Capsaicin 28-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 15689199-11 2005 This is most likely due to a feedback inhibition of nitric oxide synthases, which indicates that the c-fos response to capsaicin within TNC is mediated by NO dependent mechanisms. Capsaicin 119-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 15845081-9 2005 Although estradiol did not alter the overall number of Fos-positive nuclei, it significantly increased the number of Fos/5-HT double-labelled cells in the medial and lateral DRN. Estradiol 9-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 15845093-8 2005 We then confirmed that c-Fos was induced in 60% of relaxin 3 neurons in the NI and the expression of relaxin 3 mRNA increased significantly in the NI after water-restraint stress. Water 156-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 16004321-0 2005 [Antagonistic effects of selenium on the expression of c-fos in central nerval system of rat included by mercury contaminated rice]. Selenium 25-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 15713275-6 2005 Epidural application of picrotoxin to the rat"s M1 motor cortex induced Fos in ipsilateral dorsolateral striatum. Picrotoxin 24-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 15713275-7 2005 Animals previously given 6-hydroxydopamine (6-OHDA) injections into the ascending DA pathways had greater total numbers of cortical stimulation-induced striatal Fos-ir cells but fewer Fos-ir/PV-ir cells, compared to sham-operates. Oxidopamine 25-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-164 15713275-7 2005 Animals previously given 6-hydroxydopamine (6-OHDA) injections into the ascending DA pathways had greater total numbers of cortical stimulation-induced striatal Fos-ir cells but fewer Fos-ir/PV-ir cells, compared to sham-operates. Oxidopamine 25-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 184-187 15713275-7 2005 Animals previously given 6-hydroxydopamine (6-OHDA) injections into the ascending DA pathways had greater total numbers of cortical stimulation-induced striatal Fos-ir cells but fewer Fos-ir/PV-ir cells, compared to sham-operates. Oxidopamine 44-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-164 15713275-7 2005 Animals previously given 6-hydroxydopamine (6-OHDA) injections into the ascending DA pathways had greater total numbers of cortical stimulation-induced striatal Fos-ir cells but fewer Fos-ir/PV-ir cells, compared to sham-operates. Oxidopamine 44-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 184-187 15713275-8 2005 In a separate experiment, rats given cortical stimulation and treated with the DA D2-class antagonist eticlopride (0.10 mg/kg) exhibited fewer Fos-ir/PV-ir cells than did vehicle-treated rats. eticlopride 102-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-146 15611033-8 2005 In a parallel set of experiments, acute rehydration of 48 h water-deprived rats significantly attenuated the increased Fos immunoreactivity in PVN neurones that project to the spinal cord or rostral ventrolateral medulla. Water 60-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-122 15758184-7 2005 Furthermore, ERK1/2, when only coactivated by both IP3/Ca2+- and Homer1b/c-dependent pathways, showed the ability to phosphorylate two transcription factors, Elk-1 and cAMP response element-binding protein, and thereby facilitated c-Fos expression. Inositol 1,4,5-Trisphosphate 51-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 231-236 15702242-1 2005 The effect of acute brief seizures on neocortical c-fos expression was investigated in rats injected with 5 mg/kg 4-aminopyridine. 4-Aminopyridine 114-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 15740792-8 2005 Immunocytochemistry, histochemistry and western blot were performed to determine the effect of midazolam on formalin-induced expression of Fos protein, nicotinamide adenine dinucleotide phosphate-diaphorase (NADPH-d) and nitric oxide synthase (NOS) in chronic morphine-tolerant rats, respectively. Midazolam 95-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 15740792-8 2005 Immunocytochemistry, histochemistry and western blot were performed to determine the effect of midazolam on formalin-induced expression of Fos protein, nicotinamide adenine dinucleotide phosphate-diaphorase (NADPH-d) and nitric oxide synthase (NOS) in chronic morphine-tolerant rats, respectively. Formaldehyde 108-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 15740792-10 2005 In chronic morphine-tolerant rats, pretreatment with midazolam significantly decreased the formalin-induced expression of Fos and Fos/NADPH-d double-labeled neurons in the contralateral spinal cord and NADPH-d positive neurons in the bilateral spinal cord. Midazolam 53-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 15740792-10 2005 In chronic morphine-tolerant rats, pretreatment with midazolam significantly decreased the formalin-induced expression of Fos and Fos/NADPH-d double-labeled neurons in the contralateral spinal cord and NADPH-d positive neurons in the bilateral spinal cord. Midazolam 53-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-133 15740792-10 2005 In chronic morphine-tolerant rats, pretreatment with midazolam significantly decreased the formalin-induced expression of Fos and Fos/NADPH-d double-labeled neurons in the contralateral spinal cord and NADPH-d positive neurons in the bilateral spinal cord. Formaldehyde 91-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 15740792-10 2005 In chronic morphine-tolerant rats, pretreatment with midazolam significantly decreased the formalin-induced expression of Fos and Fos/NADPH-d double-labeled neurons in the contralateral spinal cord and NADPH-d positive neurons in the bilateral spinal cord. Formaldehyde 91-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-133 15740792-10 2005 In chronic morphine-tolerant rats, pretreatment with midazolam significantly decreased the formalin-induced expression of Fos and Fos/NADPH-d double-labeled neurons in the contralateral spinal cord and NADPH-d positive neurons in the bilateral spinal cord. Deuterium 55-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 15740792-10 2005 In chronic morphine-tolerant rats, pretreatment with midazolam significantly decreased the formalin-induced expression of Fos and Fos/NADPH-d double-labeled neurons in the contralateral spinal cord and NADPH-d positive neurons in the bilateral spinal cord. Deuterium 55-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-133 16004321-0 2005 [Antagonistic effects of selenium on the expression of c-fos in central nerval system of rat included by mercury contaminated rice]. Mercury 105-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 16004321-3 2005 The results show the Hg-contaminated rice induced significantly the expression of c-fos mRNA and c-FOS protein; selenium could antagonize mercury accumulative level in brain. Selenium 112-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 16004321-4 2005 Antagonistic effects of selenium on the expression of c-fos included by mercury and the molecule mechanism of the antagonisms between selenium and mercury was probed, too. Selenium 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 16004321-4 2005 Antagonistic effects of selenium on the expression of c-fos included by mercury and the molecule mechanism of the antagonisms between selenium and mercury was probed, too. Mercury 72-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 15880951-5 2005 MATERIAL AND METHODS: The expression of c-fos and IL-1beta was identified after injecting saline solution, 10 microg of lipopolysaccharide or 100 microg of lipopolysaccharide into the larynx of 12 rats. Sodium Chloride 90-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 15713429-0 2005 Citalopram combined with WAY 100635 inhibits ejaculation and ejaculation-related Fos immunoreactivity. Citalopram 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-84 15713429-0 2005 Citalopram combined with WAY 100635 inhibits ejaculation and ejaculation-related Fos immunoreactivity. N-(2-(4-(2-methoxyphenyl)-1-piperazinyl)ethyl)-N-(2-pyridinyl)cyclohexanecarboxamide 25-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-84 15713429-8 2005 Brain sites associated with ejaculation showed reduced Fos-immunoreactivity in rats treated with both citalopram and WAY 100635. Citalopram 102-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 15713429-9 2005 Citalopram reduced Fos-immunoreactivity in the arcuate hypothalamic nucleus, an area that might link serotonergic neurotransmission to ejaculation. Citalopram 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 15671265-11 2005 Western blot analysis showed that the c-fos protein level increased in the nuclear fraction after a 6-hour exposure to light, but was decreased in PBN-treated rats. phenyl-N-tert-butylnitrone 147-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 15671265-12 2005 CONCLUSIONS: Inhibition of c-fos activation by PBN may be the key event in protection. phenyl-N-tert-butylnitrone 47-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 15671265-13 2005 The involvement of oxygen free radicals has been suggested in c-fos activation and the action of PBN could be through its antioxidant activity. oxygen free radicals 19-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 15680264-0 2005 AM404, an inhibitor of anandamide reuptake decreases Fos-immunoreactivity in the spinal cord of neuropathic rats after non-noxious stimulation. N-(4-hydroxyphenyl)arachidonylamide 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 15686494-0 2005 Activation of c-fos by lipopolysaccharide in glial cells via p38 mitogen-activated protein kinase-dependent activation of serum or cyclic AMP/calcium response element. Cyclic AMP 131-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 15686494-0 2005 Activation of c-fos by lipopolysaccharide in glial cells via p38 mitogen-activated protein kinase-dependent activation of serum or cyclic AMP/calcium response element. Calcium 142-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 15649440-0 2005 Neural segregation of Fos-protein distribution in the brain following freezing and escape behaviors induced by injections of either glutamate or NMDA into the dorsal periaqueductal gray of rats. Glutamic Acid 132-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-25 15649440-0 2005 Neural segregation of Fos-protein distribution in the brain following freezing and escape behaviors induced by injections of either glutamate or NMDA into the dorsal periaqueductal gray of rats. N-Methylaspartate 145-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-25 15649440-1 2005 Freezing and escape responses induced by gradual increases in the intensity of the electrical current applied to dorsal regions of the periaqueductal gray (dPAG) cause a distinct pattern of Fos distribution in the brain. dpag 156-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 190-193 15649440-4 2005 To examine further this issue we measured Fos protein expression in brain areas activated by stimulation of the dPAG with glutamate (5 nmol/0.2 microL) and N-methyl-D-aspartate (NMDA) at doses that provoke either freezing (4 nmol/0.2 microL) or escape (7 nmol/0.2 microL) responses, respectively. dpag 112-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 15649440-4 2005 To examine further this issue we measured Fos protein expression in brain areas activated by stimulation of the dPAG with glutamate (5 nmol/0.2 microL) and N-methyl-D-aspartate (NMDA) at doses that provoke either freezing (4 nmol/0.2 microL) or escape (7 nmol/0.2 microL) responses, respectively. Glutamic Acid 122-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 15649440-5 2005 The results showed that glutamate-induced freezing caused a selective increase in Fos expression in the superior and inferior colliculi as well as in the laterodorsal nucleus of the thalamus. Glutamic Acid 24-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 15649440-6 2005 On the other hand, NMDA-induced escape led to widespread increases in Fos labeling in almost all structures studied. N-Methylaspartate 19-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 15649440-7 2005 Differently from glutamate, NMDA at doses provoking freezing caused significant increase of Fos labeling in the dPAG and CnF. N-Methylaspartate 28-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 15458969-5 2005 The number of c-Fos-positive cells in the SON was significantly increased after 24 and 48 h of water deprivation. Water 95-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 15458969-7 2005 Water deprivation significantly increased c-Fos staining in both the OVLT and the MnPO, but c-Fos staining was not altered by rehydration. Water 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 15458969-12 2005 We conclude that water deprivation and rehydration differentially affect c-Fos and FosB staining in a region-dependent manner. Water 17-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 16180654-0 2005 Intrathecal grafting of porcine chromaffin cells reduces formalin-evoked c-Fos expression in the rat spinal cord. Formaldehyde 57-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 16180654-4 2005 The goal of the present study was to confirm porcine chromaffin cell analgesic effects at the molecular level by evaluating neural activity as reflected by spinal cord c-Fos protein expression. chromaffin 53-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-173 16180654-5 2005 To this end, the expression of c-Fos in response to intraplantar formalin injection was evaluated in animals following intrathecal grafting of 10(6) porcine or bovine chromaffin cells. Formaldehyde 65-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 16180654-9 2005 The present study demonstrates that both xenogeneic porcine and bovine chromaffin cells transplanted into the spinal subarachnoid space of the rat can suppress formalin-evoked c-Fos expression equally, in parallel with suppression of nociceptive behaviors in the tonic phase of the test. chromaffin 71-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 176-181 16180654-9 2005 The present study demonstrates that both xenogeneic porcine and bovine chromaffin cells transplanted into the spinal subarachnoid space of the rat can suppress formalin-evoked c-Fos expression equally, in parallel with suppression of nociceptive behaviors in the tonic phase of the test. Formaldehyde 160-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 176-181 15629133-4 2005 NaHS alleviated the neuronal damage of recurrent FS rats, decreased the expression of c-fos, and inhibited MFS obviously. sodium bisulfide 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 15593338-3 2005 Bilateral ibotenic acid lesions of the rat mPFC enhanced the number of Fos-immunoreactive cells seen in the PVN after exposure to the psychological stressor, air puff. Ibotenic Acid 10-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 15619548-5 2005 ), and simultaneously inhibited pentylenetetrazol-stimulated c-Fos expression in some areas of the hippocampus. Pentylenetetrazole 32-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 15661368-1 2005 The purpose of this study was to determine whether activation of ATP-sensitive K+ (KATP) channels with diazoxide (DIZ) is able to prevent the cleavage of cytosolic mu-calpain and abrogate the elevation of nuclear c-Fos and c-Jun protein (c-Fos, c-Jun) expressions after hypoxic-ischemia (HI) in brain. Diazoxide 103-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-218 15661368-1 2005 The purpose of this study was to determine whether activation of ATP-sensitive K+ (KATP) channels with diazoxide (DIZ) is able to prevent the cleavage of cytosolic mu-calpain and abrogate the elevation of nuclear c-Fos and c-Jun protein (c-Fos, c-Jun) expressions after hypoxic-ischemia (HI) in brain. Diazoxide 103-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 238-243 15486230-7 2005 The induction of c-fos expression in glutamic acid decarboxylase67 immunostained neurons in the preoptic area suggests activation of the secretion of gamma-aminobutyric acid in response to intracoerulear administration of CRH; 17beta-estradiol further increased the percentage of glutamic acid decarboxylase67-positive neurons that expressed fos and augmented suppression of LH pulses. gamma-Aminobutyric Acid 150-173 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 15486230-7 2005 The induction of c-fos expression in glutamic acid decarboxylase67 immunostained neurons in the preoptic area suggests activation of the secretion of gamma-aminobutyric acid in response to intracoerulear administration of CRH; 17beta-estradiol further increased the percentage of glutamic acid decarboxylase67-positive neurons that expressed fos and augmented suppression of LH pulses. gamma-Aminobutyric Acid 150-173 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 15486230-7 2005 The induction of c-fos expression in glutamic acid decarboxylase67 immunostained neurons in the preoptic area suggests activation of the secretion of gamma-aminobutyric acid in response to intracoerulear administration of CRH; 17beta-estradiol further increased the percentage of glutamic acid decarboxylase67-positive neurons that expressed fos and augmented suppression of LH pulses. Estradiol 227-243 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 15486230-7 2005 The induction of c-fos expression in glutamic acid decarboxylase67 immunostained neurons in the preoptic area suggests activation of the secretion of gamma-aminobutyric acid in response to intracoerulear administration of CRH; 17beta-estradiol further increased the percentage of glutamic acid decarboxylase67-positive neurons that expressed fos and augmented suppression of LH pulses. Estradiol 227-243 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 15486230-7 2005 The induction of c-fos expression in glutamic acid decarboxylase67 immunostained neurons in the preoptic area suggests activation of the secretion of gamma-aminobutyric acid in response to intracoerulear administration of CRH; 17beta-estradiol further increased the percentage of glutamic acid decarboxylase67-positive neurons that expressed fos and augmented suppression of LH pulses. Luteinizing Hormone 375-377 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 15486230-7 2005 The induction of c-fos expression in glutamic acid decarboxylase67 immunostained neurons in the preoptic area suggests activation of the secretion of gamma-aminobutyric acid in response to intracoerulear administration of CRH; 17beta-estradiol further increased the percentage of glutamic acid decarboxylase67-positive neurons that expressed fos and augmented suppression of LH pulses. Luteinizing Hormone 375-377 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 15680264-0 2005 AM404, an inhibitor of anandamide reuptake decreases Fos-immunoreactivity in the spinal cord of neuropathic rats after non-noxious stimulation. anandamide 23-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 15680264-2 2005 In this study we evaluated the effects of administrating N-(4-hydroxyphenyl)-5Z,8Z,11Z,14Z-eicosatetraenamide (AM404), an inhibitor of anandamide reuptake and monitoring the expression of c-fos, a marker of activated neurons in an experimental model of neuropathic pain (sciatic nerve tying). N-(4-hydroxyphenyl)arachidonylamide 111-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 188-193 15680264-5 2005 AM404 significantly reduced Fos induction in tied animals. N-(4-hydroxyphenyl)arachidonylamide 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 15943228-6 2005 In comparison to sham-operated animals, application of veratrine is associated with a statistically significant increase (P < 0.01) of the mean number of Fos-ir neurons in the Sol and CVL, but not in the RVL. Veratrine 55-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-160 15859433-0 2005 [Expression of c-FOS protein in rat hippocampus by inducing of mercury contaminated rice]. Mercury 63-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 15859433-2 2005 The results show the mercury pollutes rice induced significantly the expression of c-FOS protein in hippccampus;the antagonisis between selenium and mercury on the exposure process. Mercury 21-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 15859433-2 2005 The results show the mercury pollutes rice induced significantly the expression of c-FOS protein in hippccampus;the antagonisis between selenium and mercury on the exposure process. Selenium 136-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 15859433-2 2005 The results show the mercury pollutes rice induced significantly the expression of c-FOS protein in hippccampus;the antagonisis between selenium and mercury on the exposure process. Mercury 149-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 15859433-3 2005 It is suggested that c-fos can be used as an effective index of detecting and assessing neurotoxicology of mercury polluted areas. Mercury 107-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 16295883-0 2005 Effects of mercury contaminated rice from typical chemical plant area in China on nitric oxide changes and c-fos expression of rats brain. Mercury 11-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 16295883-6 2005 The results showed the neural transmitter NO and NOS in brain were significantly change between exposure groups and control group; the mercury polluted rice induced significantly the expression of c-fos mRNA; the c-FOS positive cells in hippocampus and cortex of exposure groups were significant different from control group (p < 0.01). Mercury 135-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 197-202 16295883-6 2005 The results showed the neural transmitter NO and NOS in brain were significantly change between exposure groups and control group; the mercury polluted rice induced significantly the expression of c-fos mRNA; the c-FOS positive cells in hippocampus and cortex of exposure groups were significant different from control group (p < 0.01). Mercury 135-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-218 16295883-7 2005 It could be concluded that nitric oxide was involved in mercury contaminated rice induced immediate early gene c-fos expressions in the rat brain. Nitric Oxide 27-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 16295883-7 2005 It could be concluded that nitric oxide was involved in mercury contaminated rice induced immediate early gene c-fos expressions in the rat brain. Mercury 56-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 16463674-6 2005 It was found that Ang II could increase the c-fos protein expression, which could be inhibited by CsA in a dose-dependent manner. Cyclosporine 98-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 15673443-7 2005 In keeping with our hypothesis, amphetamine- or IL-1beta-induced c-fos and zif-268 mRNA were significantly decreased in the CEAl and BSTov under conditions of loud noise or restraint stress compared with control conditions. Amphetamine 32-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 15663473-0 2005 Involvement of 3",5"-cyclic adenosine monophosphate-dependent protein kinase in regulation of Fos expression and tyrosine hydroxylase levels during morphine withdrawal in the hypothalamic paraventricular nucleus and medulla oblongata catecholaminergic cell groups. Morphine 148-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 15663473-6 2005 Morphine withdrawal induced expression of Fos in the PVN and NTS/VLM, indicating an activation of neurones in those nuclei. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 15663473-8 2005 When the selective PKA inhibitor HA-1004 was infused it greatly diminished the Fos expression observed in morphine-withdrawn rats. N-(2-guanidinoethyl)-5-isoquinolinesulfonamide 33-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 15663473-8 2005 When the selective PKA inhibitor HA-1004 was infused it greatly diminished the Fos expression observed in morphine-withdrawn rats. Morphine 106-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 16463674-9 2005 It was concluded that CsA can suppress the Ang II-induced c-fos protein expression and [Ca2+]i elevation in single cardiomyocyte, which might play a role in the prevention of Ang II-induced cardiomyocyte hypertrophy by CsA. Cyclosporine 22-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 16463674-9 2005 It was concluded that CsA can suppress the Ang II-induced c-fos protein expression and [Ca2+]i elevation in single cardiomyocyte, which might play a role in the prevention of Ang II-induced cardiomyocyte hypertrophy by CsA. Cyclosporine 219-222 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 16401911-0 2005 Induction of Fos immunoreactivity labeling in rat forebrain metabolic loci by caudal fourth ventricular infusion of the monocarboxylate transporter inhibitor, alpha-cyano-4-hydroxycinnamic acid. alpha-cyano-4-hydroxycinnamate 159-193 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 16019152-0 2005 Effects of chronic paroxetine pretreatment on (+/-)-8-hydroxy-2-(di-n-propyl-amino)tetralin induced c-fos expression following sexual behavior. Paroxetine 19-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 16401911-1 2005 Caudal fourth ventricular (CV4) infusion of the monocarboxylate transporter inhibitor, alpha-cyano-4-hydroxycinnamic acid (4CIN), causes hyperglycemia coincident with Fos expression in the hindbrain nucleus tractus solitarius, a rare central source of metabolic deficit signaling. alpha-cyano-4-hydroxycinnamate 87-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 167-170 16401911-1 2005 Caudal fourth ventricular (CV4) infusion of the monocarboxylate transporter inhibitor, alpha-cyano-4-hydroxycinnamic acid (4CIN), causes hyperglycemia coincident with Fos expression in the hindbrain nucleus tractus solitarius, a rare central source of metabolic deficit signaling. alpha-cyano-4-hydroxycinnamate 123-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 167-170 16401911-3 2005 Two hours after CV4 infusion of graded doses of 4CIN or vehicle alone, adult female rats were sacrificed by transcardial perfusion and sections through the telencephalic and diencephalic metabolic loci were processed for Fos immunoreactivity (-ir). alpha-cyano-4-hydroxycinnamate 48-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 221-224 16401911-4 2005 Fos labeling of the hypothalamic paraventricular (PVH), dorsomedial (DMH), and ventromedial (VMH) nuclei was significantly elevated, relative to the vehicle-treated controls, in response to the lowest dose of 4CIN, e.g. 10 microg/animal. Dimenhydrinate 69-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 16401911-4 2005 Fos labeling of the hypothalamic paraventricular (PVH), dorsomedial (DMH), and ventromedial (VMH) nuclei was significantly elevated, relative to the vehicle-treated controls, in response to the lowest dose of 4CIN, e.g. 10 microg/animal. alpha-cyano-4-hydroxycinnamate 209-213 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 16401911-5 2005 Treatment with higher doses of 4CIN (25 or 50 microg) further augmented numbers of Fos-ir-positive neurons in these structures, and also elicited staining of the bed nuclei of the stria terminalis (BST), medial preoptic (MPN), arcuate (ARH), supraoptic (SO), and anterior hypothalamic nuclei (AHN), and lateral hypothalamic area (LHA). alpha-cyano-4-hydroxycinnamate 31-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 16401911-6 2005 Mean numbers of Fos-immunolabeled neurons in the ARH, DMH, LHA, AHN, MPN, and SO were not different between animals infused with 25 versus 50 microg 4CIN, whereas neuronal labeling in the VMH, BST, and PVH was significantly greater in the high- versus the middle-dose groups. Dimenhydrinate 54-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 16401911-7 2005 The present data show that pharmacological inhibition of lactate uptake within the caudal hindbrain results in dose-dependent neuronal Fos immunoexpression within characterized forebrain components of the central metabolic circuitry, and that these patterns of neuronal transcriptional activation parallel observed drug effects on blood glucose levels. Lactic Acid 57-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-138 16084651-4 2005 Indeed, systemic as well as local administrations of the serotonin agonist quipazine in the region of the suprachiasmatic nucleus mimic the effects of light on the circadian system of rats, i.e. they induce phase-advances of the locomotor activity rhythm as well as c-FOS expression in the suprachiasmatic nucleus during late subjective night. Serotonin 57-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 268-271 16084651-4 2005 Indeed, systemic as well as local administrations of the serotonin agonist quipazine in the region of the suprachiasmatic nucleus mimic the effects of light on the circadian system of rats, i.e. they induce phase-advances of the locomotor activity rhythm as well as c-FOS expression in the suprachiasmatic nucleus during late subjective night. Quipazine 75-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 268-271 16084664-6 2005 c-fos and NGFI-B were also upregulated by nicotine, but not in an age-related manner. Nicotine 42-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 16084664-7 2005 In contrast, nicotine induced less arc, c-fos, and NGFI-B expression in the somatosensory cortex of adolescents compared with adults. Nicotine 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 16019152-0 2005 Effects of chronic paroxetine pretreatment on (+/-)-8-hydroxy-2-(di-n-propyl-amino)tetralin induced c-fos expression following sexual behavior. 8-Hydroxy-2-(di-n-propylamino)tetralin 46-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 16019152-10 2005 Chronic treatment with paroxetine reduced Fos-immunoreactivity in the locus coeruleus, and prevented the increase in Fos-immunoreactive neurons induced by 8-OH-DPAT in the oxytocinergic magnocellular part of the paraventricular nucleus as well as in the locus coeruleus. Paroxetine 23-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 16019152-10 2005 Chronic treatment with paroxetine reduced Fos-immunoreactivity in the locus coeruleus, and prevented the increase in Fos-immunoreactive neurons induced by 8-OH-DPAT in the oxytocinergic magnocellular part of the paraventricular nucleus as well as in the locus coeruleus. Paroxetine 23-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 16019152-10 2005 Chronic treatment with paroxetine reduced Fos-immunoreactivity in the locus coeruleus, and prevented the increase in Fos-immunoreactive neurons induced by 8-OH-DPAT in the oxytocinergic magnocellular part of the paraventricular nucleus as well as in the locus coeruleus. 8-Hydroxy-2-(di-n-propylamino)tetralin 155-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 16112474-0 2005 Preference for cocaine- versus pup-associated cues differentially activates neurons expressing either Fos or cocaine- and amphetamine-regulated transcript in lactating, maternal rodents. Cocaine 15-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 16019152-12 2005 Chronic paroxetine treatment and 8-OH-DPAT changed c-fos expression in a number of other brain areas, indicating that Fos-immunohistochemistry is a useful tool to find locations where selective serotonin reuptake inhibitors and 8-OH-DPAT exert their effects. Paroxetine 8-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 16112474-4 2005 Dams that preferred the cocaine-associated cues had more c-Fos positive neurons in medial prefrontal cortex, nucleus accumbens, and basolateral nucleus of amygdala than pup-associated cue preferring dams or control. Cocaine 24-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 16019152-12 2005 Chronic paroxetine treatment and 8-OH-DPAT changed c-fos expression in a number of other brain areas, indicating that Fos-immunohistochemistry is a useful tool to find locations where selective serotonin reuptake inhibitors and 8-OH-DPAT exert their effects. Paroxetine 8-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 16019152-12 2005 Chronic paroxetine treatment and 8-OH-DPAT changed c-fos expression in a number of other brain areas, indicating that Fos-immunohistochemistry is a useful tool to find locations where selective serotonin reuptake inhibitors and 8-OH-DPAT exert their effects. 8-Hydroxy-2-(di-n-propylamino)tetralin 33-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 16019152-12 2005 Chronic paroxetine treatment and 8-OH-DPAT changed c-fos expression in a number of other brain areas, indicating that Fos-immunohistochemistry is a useful tool to find locations where selective serotonin reuptake inhibitors and 8-OH-DPAT exert their effects. 8-Hydroxy-2-(di-n-propylamino)tetralin 33-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 16019152-12 2005 Chronic paroxetine treatment and 8-OH-DPAT changed c-fos expression in a number of other brain areas, indicating that Fos-immunohistochemistry is a useful tool to find locations where selective serotonin reuptake inhibitors and 8-OH-DPAT exert their effects. 8-Hydroxy-2-(di-n-propylamino)tetralin 228-237 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 16019152-12 2005 Chronic paroxetine treatment and 8-OH-DPAT changed c-fos expression in a number of other brain areas, indicating that Fos-immunohistochemistry is a useful tool to find locations where selective serotonin reuptake inhibitors and 8-OH-DPAT exert their effects. 8-Hydroxy-2-(di-n-propylamino)tetralin 228-237 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 15590158-10 2005 Double-labeling studies revealed that of the c-Fos-positive neurons responsive to hypertonic NaCl, 52%, 41%, and 3% exhibited immunoreactivity for Glu, SP, and CGRP, respectively. Glutamic Acid 147-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 16183203-0 2005 Glutamatergic stimulation of the medial amygdala induces steroid dependent c-fos expression within forebrain nuclei responsive to mating stimulation. Steroids 57-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 15590158-12 2005 In addition, 44%, 38%, and 8% of 5-HT-stimulated and 30%, 32%, and 5% of maltose-stimulated c-Fos-positive neurons exhibited, respectively, Glu, SP, and CGRP immunoreactivity. Maltose 73-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 15590158-12 2005 In addition, 44%, 38%, and 8% of 5-HT-stimulated and 30%, 32%, and 5% of maltose-stimulated c-Fos-positive neurons exhibited, respectively, Glu, SP, and CGRP immunoreactivity. Glutamic Acid 140-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 15664699-1 2005 There is evidence that metyrapone (MET), apart from its inhibition of 11-beta steroid hydroxylation, may exert some stress-like effects in the brain, including the activation of the hypothalamic-pituitary-adrenal (HPA) axis and the induction of c-fos. Metyrapone 23-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 245-250 15857696-6 2005 The percentage of Fos-positive cells expressing NK1 immunoreaction did not change in monoarthritics but that of Fos cells with GABA(B) immunoreaction was lower in these animals. gamma-Aminobutyric Acid 127-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 15708488-0 2005 Effects of injections of 8-hydroxy-2-(di-n-propylamino)tetralin or muscimol in the median raphe nucleus on c-fos mRNA in the rat brain. 8-Hydroxy-2-(di-n-propylamino)tetralin 25-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 15708488-0 2005 Effects of injections of 8-hydroxy-2-(di-n-propylamino)tetralin or muscimol in the median raphe nucleus on c-fos mRNA in the rat brain. Muscimol 67-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 15708488-10 2005 In the ventral hippocampus, only 8-OH-DPAT increased c-fos, while in the basolateral nucleus of the amygdala and locus coeruleus, it was increased only by muscimol. 8-Hydroxy-2-(di-n-propylamino)tetralin 33-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 15857711-7 2005 Carrageenan induced conspicuous expression of c-fos-like immunoreactivity (FLI) in the spinal dorsal horn of segments L4-5. Carrageenan 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 15896914-5 2005 In Sal- or Bl-Sap-treated controls, high intensity stimulation induced c-Fos expression in neurons throughout the full extent of ipsilateral superficial layers of the Vc (VcI/II), magnocellular zone of the Vc (VcIII/IV) and the dorsal or dorsomedial subdivisions of the rostral TSN above the obex (trigeminal principal, oral (Vo) and interpolar nuclei). bl-sap 11-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 15893646-6 2005 Fos immunohistochemistry was used to identify brainstem neurons activated by a brief (90 s) intraoral infusion of a small volume (90 microl, 0.2M) of sucrose or a salt solution (0.1 M ammonium chloride) in 10-day-old rat pups. Sucrose 150-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 15893646-6 2005 Fos immunohistochemistry was used to identify brainstem neurons activated by a brief (90 s) intraoral infusion of a small volume (90 microl, 0.2M) of sucrose or a salt solution (0.1 M ammonium chloride) in 10-day-old rat pups. salt solution 163-176 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 15893646-6 2005 Fos immunohistochemistry was used to identify brainstem neurons activated by a brief (90 s) intraoral infusion of a small volume (90 microl, 0.2M) of sucrose or a salt solution (0.1 M ammonium chloride) in 10-day-old rat pups. Ammonium Chloride 184-201 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 15893646-7 2005 Compared with control groups (intact, cannula, distilled water), both sucrose and ammonium chloride induced Fos expression in the rostral nucleus tractus solitarius, the first relay in the ascending gustatory pathway. Sucrose 70-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 15893646-7 2005 Compared with control groups (intact, cannula, distilled water), both sucrose and ammonium chloride induced Fos expression in the rostral nucleus tractus solitarius, the first relay in the ascending gustatory pathway. Ammonium Chloride 82-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 15893646-8 2005 Sucrose also elicited Fos expression in several brainstem areas associated with centrally mediated analgesia, including the periaqueductal gray and the nucleus raphe magnus. Sucrose 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-25 15896914-0 2005 Elimination of neurokinin-1 receptor neurons in caudal nucleus reverses the effects of systemic bicuculline on c-Fos expression in rat trigeminal sensory nucleus: I. Bicuculline 96-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 15896914-3 2005 This study examines the effect of intra cisterna magna injection of substance P (SP) conjugated to saporin (SP-Sap; 5 microM, 5 microl) [with/without systemic administration of bicuculline] on c-Fos expression in the trigeminal sensory nucleus (TSN) induced 2 h after 10 min repetitive electrical stimulation of the trigeminal ganglion (TG) at high intensity (1.0 mA, 5 Hz, 5 ms) in the urethane-anesthetized rat. sp-sap 108-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-198 15939543-8 2005 In an immunohistochemical study for c-Fos protein, naloxone-precipitated morphine withdrawal dramatically induced c-Fos-immunoreactive neurons in the capsular part of the Ce, and the lateral and medial divisions of the BST. Naloxone 51-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 15939543-8 2005 In an immunohistochemical study for c-Fos protein, naloxone-precipitated morphine withdrawal dramatically induced c-Fos-immunoreactive neurons in the capsular part of the Ce, and the lateral and medial divisions of the BST. Naloxone 51-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 15939543-8 2005 In an immunohistochemical study for c-Fos protein, naloxone-precipitated morphine withdrawal dramatically induced c-Fos-immunoreactive neurons in the capsular part of the Ce, and the lateral and medial divisions of the BST. Morphine 73-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 15896914-7 2005 decreased the numbers of c-Fos-immunopositive neurons in the VcI/II, VcIII/IV and Vo in Sal- or Bl-Sap-treated controls. Sodium Chloride 88-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 15939543-8 2005 In an immunohistochemical study for c-Fos protein, naloxone-precipitated morphine withdrawal dramatically induced c-Fos-immunoreactive neurons in the capsular part of the Ce, and the lateral and medial divisions of the BST. Morphine 73-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 15939543-9 2005 Bilateral excitotoxic lesion of the Ce reduced the number of morphine withdrawal-induced c-Fos-immunoreactive neurons in the lateral and medial BST, with significant decreases in the posterior, ventral and juxtacapsular parts of lateral division, and anterior part of the medial division, but not in the ventral part of the medial division of the BST. Morphine 61-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 15896914-8 2005 In contrast, high intensity stimulation induced less c-Fos-immunopositive neurons in the VcI/II and Vo of rats treated with SP-Sap compared with those in Sal- or Bl-Sap-treated controls. sp-sap 124-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 15896914-9 2005 In SP-Sap-treated rats preadministered with bicuculline, the numbers of c-Fos-immunopositive neurons in the VcI/II and Vo were increased compared with the SP-Sap-treated rats preadministered with Sal. sp-sap 3-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 15896914-9 2005 In SP-Sap-treated rats preadministered with bicuculline, the numbers of c-Fos-immunopositive neurons in the VcI/II and Vo were increased compared with the SP-Sap-treated rats preadministered with Sal. Bicuculline 44-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 15953686-0 2005 Acute fluoxetine administration differentially affects brain C-Fos expression in fasted and refed rats. Fluoxetine 6-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 15896914-9 2005 In SP-Sap-treated rats preadministered with bicuculline, the numbers of c-Fos-immunopositive neurons in the VcI/II and Vo were increased compared with the SP-Sap-treated rats preadministered with Sal. Sodium Chloride 196-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 15953686-1 2005 In the present study we investigated the effect of acute fluoxetine administration on the expression of c-Fos in the rat brain under two different metabolic conditions: fed and fasting states. Fluoxetine 57-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 15953686-9 2005 These data indicate that the metabolic condition of the animals significantly modifies fluoxetine-induced brain c-Fos expression, suggesting that visceral and behavioral fluoxetine effects may be influenced by the metabolic state of the individual. Fluoxetine 87-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 15953686-7 2005 Both in fasting and fed states, fluoxetine-treated animals presented a significant increase in c-Fos expression in hypothalamic areas, limbic structures, circumventricular areas, and in mesencephalic and rhomboencephalic regions, as compared with saline-treated controls. Fluoxetine 32-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 15953686-8 2005 The quantitative comparison of data obtained from fasted and fed animals showed that fasted rats treated with fluoxetine presented a higher c-Fos expression in the ventromedial hypothalamus and the paraventricular nuclei compared with the fed group, while in fluoxetine-treated fed rats c-Fos expression was higher in the arcuate nuclei, medial amygdala, locus coeruleus and dorsal raphe nuclei, as compared with fasted, fluoxetine-treated animals. Fluoxetine 110-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 15953686-8 2005 The quantitative comparison of data obtained from fasted and fed animals showed that fasted rats treated with fluoxetine presented a higher c-Fos expression in the ventromedial hypothalamus and the paraventricular nuclei compared with the fed group, while in fluoxetine-treated fed rats c-Fos expression was higher in the arcuate nuclei, medial amygdala, locus coeruleus and dorsal raphe nuclei, as compared with fasted, fluoxetine-treated animals. Fluoxetine 110-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 287-292 15953686-9 2005 These data indicate that the metabolic condition of the animals significantly modifies fluoxetine-induced brain c-Fos expression, suggesting that visceral and behavioral fluoxetine effects may be influenced by the metabolic state of the individual. Fluoxetine 170-180 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 15609068-0 2005 Contrasting Fos expression induced by acute reboxetine and fluoxetine in the rat forebrain: neuroanatomical substrates for the antidepressant effect. Reboxetine 44-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-15 15668729-5 2005 Prior to the induction of GRP78, lithium treatment alone increased the expression of c-Fos whose induction by ER stress is necessary for GRP78 induction. Lithium 33-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 15609068-0 2005 Contrasting Fos expression induced by acute reboxetine and fluoxetine in the rat forebrain: neuroanatomical substrates for the antidepressant effect. Fluoxetine 59-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-15 15609068-7 2005 RESULTS: The shell of the nucleus accumbens was the only region in which both fluoxetine and reboxetine equally increased Fos-ir expression. Fluoxetine 78-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 15609068-7 2005 RESULTS: The shell of the nucleus accumbens was the only region in which both fluoxetine and reboxetine equally increased Fos-ir expression. Reboxetine 93-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 15609068-8 2005 Fluoxetine particularly induced Fos-ir in the central nucleus of the amygdala. Fluoxetine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 15609068-9 2005 In contrast, reboxetine induced Fos-ir in the cingulate cortex area 3 and the lateral orbital cortex. Reboxetine 13-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 15769397-9 2005 CONCLUSION: Hypoxia induced expression of c-fos and c-jun genes, which might play an vital role in the early stage of PASMCs proliferation. pasmcs 118-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 15588731-9 2004 Morphine withdrawal also induced an increase in the Fos expression, which indicates an activation of cardiac cellular activity. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 15601931-12 2004 Finally, intravesical instillation of anandamide (50 microm) increased c-fos expression in the spinal cord, which was reduced by capsazepine or by resiniferatoxin pretreatment. anandamide 38-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 15525283-7 2004 In contrast, rats that had received muscimol injections into their mPFC and were subsequently restrained exhibited an increase in the number of Fos-positive cells in the DMH, medial amygdala, and medial nucleus tractus solitarius as compared to vehicle-injected rats that experienced restraint stress. Muscimol 36-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-147 15601931-12 2004 Finally, intravesical instillation of anandamide (50 microm) increased c-fos expression in the spinal cord, which was reduced by capsazepine or by resiniferatoxin pretreatment. capsazepine 129-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 15601931-12 2004 Finally, intravesical instillation of anandamide (50 microm) increased c-fos expression in the spinal cord, which was reduced by capsazepine or by resiniferatoxin pretreatment. resiniferatoxin 147-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 15601952-6 2004 Fos immunohistochemistry, however, revealed notable differences in the pattern of UcnI-induced activation between intact and CD rats. Cadmium 125-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 15598139-4 2004 Following exposure to a mobile intruder, Fos expression was lower in the medial preoptic area and the bed nucleus of the stria terminalis in L-NAME-treated rats than in controls but was lower in the medial amygdala only following exposure to an anaesthetized intruder. NG-Nitroarginine Methyl Ester 141-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 15672679-7 2004 Rats were treated intraperitoneally with a NMDA-receptor antagonist MK801 and activation of brain areas was detected by monitoring the expression of c-fos mRNA by using in situ hybridization. Dizocilpine Maleate 68-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 149-154 15672679-9 2004 MK801 induced c-fos mRNA expression of in the retrosplenial, entorhinal, and prefrontal cortices. Dizocilpine Maleate 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 15672679-14 2004 NMDA-receptor antagonist ketamine increased dopamine release in the parietal cortex, in the region where NMDA-receptor antagonist increased c-fos mRNA expression. Ketamine 25-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 15654138-0 2004 Pre-versus post-formalin effects of intrathecal ketamine on spinal Fos-like immunoreactivity in rats. Ketamine 48-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 15654138-1 2004 BACKGROUND & OBJECTIVES: The spinal expression of the c-Fos immediate early gene in response to formalin pain of the hind paw of rat was used as a marker of neuronal activity. Adenosine Monophosphate 12-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 15654138-1 2004 BACKGROUND & OBJECTIVES: The spinal expression of the c-Fos immediate early gene in response to formalin pain of the hind paw of rat was used as a marker of neuronal activity. Formaldehyde 100-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 15574735-6 2004 Furthermore, ERK activated through this PSD-95/Homer1b/c-dependent and Ca2+-independent pathway was able to phosphorylate the two key transcription factors Elk-1 and cAMP response element-binding protein, which further leads to facilitation of c-Fos expression. Cyclic AMP 166-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 244-249 15667452-1 2004 We examined the extent to which basal levels of corticosterone, which vary in a circadian fashion, influence the pattern of Fos protein expression in the paraventricular nucleus of the hypothalamus (PVN), the hippocampal formation and three different functional cortical areas. Corticosterone 48-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-127 15667452-9 2004 Corticosterone replacement normalized the adrenalectomy effect on Fos expression in both brain regions. Corticosterone 0-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 15667452-10 2004 Thus, Fos expression in the rat brain displays specific patterns of dependency on the permissive effects of glucorticoids, and this dependency varies between brain regions. glucorticoids 108-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 6-9 15371543-5 2004 RA-mediated phosphorylation of CREB leads to a direct stimulation of CREB-dependent transcriptional activity and to activation of the expression of genes such as c-fos, which do not contain RAREs but contain cAMP response elements (CREs) in their promoters. Tretinoin 0-2 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 15371543-5 2004 RA-mediated phosphorylation of CREB leads to a direct stimulation of CREB-dependent transcriptional activity and to activation of the expression of genes such as c-fos, which do not contain RAREs but contain cAMP response elements (CREs) in their promoters. Cyclic AMP 208-212 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 15567425-0 2004 Modulation of morphine-induced Fos-immunoreactivity by the cannabinoid receptor antagonist SR 141716. Morphine 14-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 15567425-8 2004 SR 141716 attenuated morphine-induced Fos-IR in several regions including the CPu, cortex, NAS (shell), LS, MnPO, MPO, paraventricular and dorsomedial hypothalamus, PV, basolateral amygdala, VTA, and Edinger-Westphal nucleus (EW). Morphine 21-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 15567425-10 2004 Possible behavioural and physiological implications of the interactive effects of SR 141716 on morphine-induced Fos-IR are discussed. Rimonabant 82-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 15567425-10 2004 Possible behavioural and physiological implications of the interactive effects of SR 141716 on morphine-induced Fos-IR are discussed. Morphine 95-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 15567487-0 2004 N(omega)-nitro-L-arginine methyl ester attenuates lithium-induced c-Fos, but not conditioned taste aversion, in rats. NG-Nitroarginine Methyl Ester 0-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 15567487-0 2004 N(omega)-nitro-L-arginine methyl ester attenuates lithium-induced c-Fos, but not conditioned taste aversion, in rats. Lithium 50-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 15567487-1 2004 Lithium chloride (LiCl) at doses sufficient to induce conditioned taste aversion (CTA) causes c-Fos expression in the relevant brain regions and activates the hypothalamic-pituitary-adrenal (HPA) axis. Lithium Chloride 0-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 15567487-1 2004 Lithium chloride (LiCl) at doses sufficient to induce conditioned taste aversion (CTA) causes c-Fos expression in the relevant brain regions and activates the hypothalamic-pituitary-adrenal (HPA) axis. Lithium Chloride 18-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 15567487-4 2004 Intraperitoneal L-NAME (30 mg/kg) given 30 min prior to LiCl significantly decreased lithium-induced c-Fos expression in the brain regions implicated in CTA learning, such as the hypothalamic paraventricular nucleus (PVN), central nucleus of amygdala (CeA), and nucleus tractus of solitarius. NG-Nitroarginine Methyl Ester 16-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 15567487-4 2004 Intraperitoneal L-NAME (30 mg/kg) given 30 min prior to LiCl significantly decreased lithium-induced c-Fos expression in the brain regions implicated in CTA learning, such as the hypothalamic paraventricular nucleus (PVN), central nucleus of amygdala (CeA), and nucleus tractus of solitarius. Lithium 85-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 15375295-0 2004 c-fos mRNA expression in rat cortical neurons during glutamate-mediated excitotoxicity. Glutamic Acid 53-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 15375295-5 2004 Glu-induced c-fos mRNA expression, under excitotoxic conditions, was inhibited by D-2-amino-5-phosphonopentanoate (AP5) but not 6-cyano-7-nitro-quinoxaline-2,3-dione (CNQX), indicating an event mediated by the NMDA subtype of Glu receptors. Glutamic Acid 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 15375295-5 2004 Glu-induced c-fos mRNA expression, under excitotoxic conditions, was inhibited by D-2-amino-5-phosphonopentanoate (AP5) but not 6-cyano-7-nitro-quinoxaline-2,3-dione (CNQX), indicating an event mediated by the NMDA subtype of Glu receptors. d-2-amino-5-phosphonopentanoate 82-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 15375295-5 2004 Glu-induced c-fos mRNA expression, under excitotoxic conditions, was inhibited by D-2-amino-5-phosphonopentanoate (AP5) but not 6-cyano-7-nitro-quinoxaline-2,3-dione (CNQX), indicating an event mediated by the NMDA subtype of Glu receptors. 2-amino-5-phosphopentanoic acid 115-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 15375295-5 2004 Glu-induced c-fos mRNA expression, under excitotoxic conditions, was inhibited by D-2-amino-5-phosphonopentanoate (AP5) but not 6-cyano-7-nitro-quinoxaline-2,3-dione (CNQX), indicating an event mediated by the NMDA subtype of Glu receptors. 6-Cyano-7-nitroquinoxaline-2,3-dione 167-171 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 15375295-5 2004 Glu-induced c-fos mRNA expression, under excitotoxic conditions, was inhibited by D-2-amino-5-phosphonopentanoate (AP5) but not 6-cyano-7-nitro-quinoxaline-2,3-dione (CNQX), indicating an event mediated by the NMDA subtype of Glu receptors. N-Methylaspartate 210-214 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 15308614-8 2004 Dual immunocytochemical labeling for septopreoptic mu-R-ir and Fos-ir demonstrated a robust induction of Fos expression by receptor-positive neurons within discrete septopreoptic sites in response to CV4 5TG, a genomic response that was diminished by CTOP pretreatment. cv4 5tg 200-207 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 15308614-8 2004 Dual immunocytochemical labeling for septopreoptic mu-R-ir and Fos-ir demonstrated a robust induction of Fos expression by receptor-positive neurons within discrete septopreoptic sites in response to CV4 5TG, a genomic response that was diminished by CTOP pretreatment. cv4 5tg 200-207 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-108 15308614-8 2004 Dual immunocytochemical labeling for septopreoptic mu-R-ir and Fos-ir demonstrated a robust induction of Fos expression by receptor-positive neurons within discrete septopreoptic sites in response to CV4 5TG, a genomic response that was diminished by CTOP pretreatment. CTOP 251-255 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 15308614-8 2004 Dual immunocytochemical labeling for septopreoptic mu-R-ir and Fos-ir demonstrated a robust induction of Fos expression by receptor-positive neurons within discrete septopreoptic sites in response to CV4 5TG, a genomic response that was diminished by CTOP pretreatment. CTOP 251-255 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-108 15494196-0 2004 Laminar distribution of GABAA- and glycine-receptor mediated tonic inhibition in the dorsal horn of the rat lumbar spinal cord: effects of picrotoxin and strychnine on expression of Fos-like immunoreactivity. Picrotoxin 139-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 182-185 15494196-0 2004 Laminar distribution of GABAA- and glycine-receptor mediated tonic inhibition in the dorsal horn of the rat lumbar spinal cord: effects of picrotoxin and strychnine on expression of Fos-like immunoreactivity. Strychnine 154-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 182-185 15494196-6 2004 There were marked regional variations in the distribution of Fos-LI cells between picrotoxin- and strychnine-treated animals. Picrotoxin 82-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 15494196-6 2004 There were marked regional variations in the distribution of Fos-LI cells between picrotoxin- and strychnine-treated animals. Strychnine 98-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 15494196-7 2004 Picrotoxin induced a significant increase in the number of Fos-LI cells throughout the dorsal horn (lamina I-VI) while strychnine significantly elevated Fos-like immunoreactivity only in deep laminae (III-VI). Picrotoxin 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 15494196-7 2004 Picrotoxin induced a significant increase in the number of Fos-LI cells throughout the dorsal horn (lamina I-VI) while strychnine significantly elevated Fos-like immunoreactivity only in deep laminae (III-VI). Strychnine 119-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-156 15494196-8 2004 For both picrotoxin and strychnine, the increase in Fos-like immunoreactivity peaked in lamina V (at 3579+/-319 and 3649+/-375% of control, respectively; mean+/-SEM) but for picrotoxin an additional peak was observed in the outer part of lamina II (1959+/-196%). Picrotoxin 9-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 15494196-8 2004 For both picrotoxin and strychnine, the increase in Fos-like immunoreactivity peaked in lamina V (at 3579+/-319 and 3649+/-375% of control, respectively; mean+/-SEM) but for picrotoxin an additional peak was observed in the outer part of lamina II (1959+/-196%). Strychnine 24-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 15494196-8 2004 For both picrotoxin and strychnine, the increase in Fos-like immunoreactivity peaked in lamina V (at 3579+/-319 and 3649+/-375% of control, respectively; mean+/-SEM) but for picrotoxin an additional peak was observed in the outer part of lamina II (1959+/-196%). Picrotoxin 174-184 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 15860926-5 2005 While vehicle administration resulted in negligible Fos immunostaining within the NTS, 4-CIN-treated animals exhibited dose-dependent increases in mean numbers of Fos-ir- and TH-/Fos-ir-positive neurons in this structure. alpha-cyano-4-hydroxycinnamate 87-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 15860926-5 2005 While vehicle administration resulted in negligible Fos immunostaining within the NTS, 4-CIN-treated animals exhibited dose-dependent increases in mean numbers of Fos-ir- and TH-/Fos-ir-positive neurons in this structure. alpha-cyano-4-hydroxycinnamate 87-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 15860926-5 2005 While vehicle administration resulted in negligible Fos immunostaining within the NTS, 4-CIN-treated animals exhibited dose-dependent increases in mean numbers of Fos-ir- and TH-/Fos-ir-positive neurons in this structure. alpha-cyano-4-hydroxycinnamate 87-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 15489007-0 2004 Grafts of immortalized chromaffin cells bio-engineered to improve met-enkephalin release also reduce formalin-evoked c-fos expression in rat spinal cord. Formaldehyde 101-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 15489007-5 2004 In the formalin hindpaw-injection model, 15 days after subarachnoid transplant of cells, grafts of met-enkephalin and pro-enkephalin cells significantly reduced the number of formalin-evoked c-fos immunoreactive spinal neurons in the spinal cord, compared to grafts of vector-alone chromaffin cells. Formaldehyde 7-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 191-196 15489007-5 2004 In the formalin hindpaw-injection model, 15 days after subarachnoid transplant of cells, grafts of met-enkephalin and pro-enkephalin cells significantly reduced the number of formalin-evoked c-fos immunoreactive spinal neurons in the spinal cord, compared to grafts of vector-alone chromaffin cells. Formaldehyde 175-183 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 191-196 15271657-1 2004 The present study sought to determine whether water deprivation increases Fos immunoreactivity, a neuronal marker related to synaptic activation, in sympathetic-regulatory neurons of the hypothalamic paraventricular nucleus (PVN). Water 46-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 15271657-3 2004 Rats were then deprived of water but not food for 48 h. Water deprivation significantly increased the number of Fos-positive nuclei throughout the dorsal, ventrolateral, and lateral parvocellular divisions of the PVN (water deprived, 215 +/- 23 cells; control, 45 +/- 7 cells, P < 0.01). Water 56-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 15271657-3 2004 Rats were then deprived of water but not food for 48 h. Water deprivation significantly increased the number of Fos-positive nuclei throughout the dorsal, ventrolateral, and lateral parvocellular divisions of the PVN (water deprived, 215 +/- 23 cells; control, 45 +/- 7 cells, P < 0.01). Water 218-223 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 15271657-4 2004 Moreover, a significantly greater number of Fos-positive nuclei were localized in spinally projecting (11 +/- 3 vs. 2 +/- 1 cells, P < 0.025) and RVLM-projecting (45 +/- 7 vs. 7 +/- 1 cells, P < 0.025) neurons of the PVN in water-deprived vs. control rats, respectively. Water 230-235 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 15271657-7 2004 In addition, we analyzed spinally projecting neurons of the RVLM and found a significantly greater percentage were Fos positive in water-deprived rats than in control rats (26 +/- 3 vs. 3 +/- 1%, respectively; P < 0.001). Water 131-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-118 15231690-2 2004 In rat liver, in particular, retinoic acid has been shown to inhibit regeneration after partial hepatectomy, most probably through repression of the expression of c-fos and c-jun. Tretinoin 29-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-168 15711026-3 2004 We have previously demonstrated that there are many more c-Fos-expressing neurons than TIDA neurons in the arcuate nucleus, and treatment with naloxone (NAL), an opioid antagonist, activated these neurons in pregnant rats. Naloxone 143-151 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 15711026-3 2004 We have previously demonstrated that there are many more c-Fos-expressing neurons than TIDA neurons in the arcuate nucleus, and treatment with naloxone (NAL), an opioid antagonist, activated these neurons in pregnant rats. Naloxone 153-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 15711026-10 2004 The percentage of c-Fos positive TH neurons in the arcute nucleus increased in rats treated with NAL for 5 h after return of pups, but not in rats treated with NAL for 2 h. Naloxone 97-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 15525283-7 2004 In contrast, rats that had received muscimol injections into their mPFC and were subsequently restrained exhibited an increase in the number of Fos-positive cells in the DMH, medial amygdala, and medial nucleus tractus solitarius as compared to vehicle-injected rats that experienced restraint stress. 1,2-Dimethylhydrazine 170-173 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-147 15469883-8 2004 Consistent with functional alpha3 knockdown, epibatidine-induced c-Fos expression in the medial habenula was attenuated in aON-treated rats. epibatidine 45-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 15469883-8 2004 Consistent with functional alpha3 knockdown, epibatidine-induced c-Fos expression in the medial habenula was attenuated in aON-treated rats. Oligonucleotides, Antisense 123-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 15378513-3 2004 We examined the effects of lesions of mPFC catecholamine terminals on local expression of Fos after exposure to air puff, a stimulus that in the rat acts as an acute psychological stressor. Catecholamines 43-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 15178552-5 2004 Intracolonic TNBS induced c-fos mRNA expression in brain nuclei involved in the autonomic, behavioral, and neuroendocrine response to a stimulus (PVN, amygdala, locus coeruleus, parabrachial nucleus, nucleus of the solitary tract) and in circumventricular organs (lamina terminalis, subfornical organ, area postrema). Trinitrobenzenesulfonic Acid 13-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 15464754-0 2004 The pattern of brain c-fos mRNA induced by a component of fox odor, 2,5-dihydro-2,4,5-trimethylthiazoline (TMT), in rats, suggests both systemic and processive stress characteristics. 2,5-dihydro-2,4,5-trimethylthiazoline 68-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 15464754-0 2004 The pattern of brain c-fos mRNA induced by a component of fox odor, 2,5-dihydro-2,4,5-trimethylthiazoline (TMT), in rats, suggests both systemic and processive stress characteristics. 2,5-dihydro-2,4,5-trimethylthiazoline 107-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 15542722-0 2004 Fos expression associated with the discriminative stimulus effects of methamphetamine in rats. Methamphetamine 70-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 15542722-3 2004 c-Fos expression in the brains of rats trained to discriminate methamphetamine from saline was significantly increased in the nucleus accumbens (NAc) and the ventral tegmental area (VTA) as compared with the expression in the control rats that were maintained under the FR-20 schedule, but no alternation was observed in other areas including the cerebral cortex, caudate putamen, substantia nigra, hippocampus, amygdala, and habenulla. Methamphetamine 63-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 15542722-3 2004 c-Fos expression in the brains of rats trained to discriminate methamphetamine from saline was significantly increased in the nucleus accumbens (NAc) and the ventral tegmental area (VTA) as compared with the expression in the control rats that were maintained under the FR-20 schedule, but no alternation was observed in other areas including the cerebral cortex, caudate putamen, substantia nigra, hippocampus, amygdala, and habenulla. Sodium Chloride 84-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 15542722-4 2004 Methamphetamine treatment in the trained rats caused a significant increase in c-Fos expression in the VTA, and a decrease in the NAc core, as compared to saline treatment. Methamphetamine 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 15542722-5 2004 However, c-Fos expression in the NAc and VTA of rats that received chronic intermittent methamphetamine administration without discrimination training, did not differ from the expression in saline-treatment animals. Methamphetamine 88-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 15353228-0 2004 Regional differences in the expression of Fos-like immunoreactivity after central salt loading in conscious rats: modulation by endogenous vasopressin and role of the area postrema. Salts 82-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 15353228-1 2004 In this study, we examined the quantitative relationship between centrally administered hypertonic saline (HS) concentrations and the expression of Fos-like immunoreactivity (FLI) in brain regions involved in the homeostasis of body fluids. Sodium Chloride 99-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-151 15554911-0 2004 Improvement of spatial cognition with dietary docosahexaenoic acid is associated with an increase in Fos expression in rat CA1 hippocampus. Docosahexaenoic Acids 46-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-104 15554911-5 2004 The number of Fos-positive neurons in the CA1 hippocampus significantly increased in DHA-treated rats compared with control rats, demonstrating a statistically significant negative correlation with the number of reference memory errors. Docosahexaenoic Acids 85-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 15554911-6 2004 These results suggest that the DHA-induced improvement in spatial cognition is associated with increased Fos expression in the CA1 hippocampus. Docosahexaenoic Acids 31-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-108 15380491-7 2004 Fos immunoreactivity in the ACC was activated by retrieval of pain-related aversion, and this activation was significantly suppressed by preadministration of AP5, but not DNQX (P <0.001). 2-amino-5-phosphopentanoic acid 158-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 15447664-5 2004 Fos expression in the brains of rats that discriminate methamphetamine from saline was significantly increased in the nucleus accumbens (NAc) and the ventral tegmental area (VTA), but not in other areas including the cerebral cortex, caudate putamen, substantia nigra, hippocampus, amygdala and habenulla, as compared with the expression in control rats that were maintained under the FR-20 schedule. Methamphetamine 55-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 15447664-5 2004 Fos expression in the brains of rats that discriminate methamphetamine from saline was significantly increased in the nucleus accumbens (NAc) and the ventral tegmental area (VTA), but not in other areas including the cerebral cortex, caudate putamen, substantia nigra, hippocampus, amygdala and habenulla, as compared with the expression in control rats that were maintained under the FR-20 schedule. Sodium Chloride 76-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 15150532-0 2004 The neurotensin agonist PD149163 increases Fos expression in the prefrontal cortex of the rat. PD149163 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 15150532-7 2004 Systemic administration of PD149163 increased overall Fos expression in the PFC, but not in the dorsal striatum. PD149163 27-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 15150532-8 2004 PD149163 induced Fos in PFC interneurons, as defined by the presence of calcium-binding proteins, and in pyramidal cells. PD149163 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 15150532-8 2004 PD149163 induced Fos in PFC interneurons, as defined by the presence of calcium-binding proteins, and in pyramidal cells. Calcium 72-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 15150532-9 2004 Pretreatment with the high-affinity neurotensin antagonist, SR48692, blocked neurotensin agonist-induced Fos expression. SR 48692 60-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-108 15482585-0 2004 Long-term suppression of methamphetamine-induced c-Fos expression in rat striatum by the injection of c-fos antisense oligodeoxynucleotides absorbed in water-absorbent polymer. Methamphetamine 25-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 15482585-0 2004 Long-term suppression of methamphetamine-induced c-Fos expression in rat striatum by the injection of c-fos antisense oligodeoxynucleotides absorbed in water-absorbent polymer. Methamphetamine 25-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 15482585-0 2004 Long-term suppression of methamphetamine-induced c-Fos expression in rat striatum by the injection of c-fos antisense oligodeoxynucleotides absorbed in water-absorbent polymer. Water 152-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 15482585-0 2004 Long-term suppression of methamphetamine-induced c-Fos expression in rat striatum by the injection of c-fos antisense oligodeoxynucleotides absorbed in water-absorbent polymer. Water 152-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 15482585-3 2004 The expression of c-Fos-immunoreactivity induced by methamphetamine (6 mg/kg, intraperitoneally) around the injection site was suppressed until 5 days after injection. Methamphetamine 52-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 15482585-4 2004 Using this method, it was observed that unilateral injection with c-fos antisense ODN into the rat striatum caused robust ipsilateral rotations after methamphetamine challenge 4 days post injection. Methamphetamine 150-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 15175841-0 2004 Withdrawal-induced c-Fos expression in the rat centromedial amygdala 24 h following a single morphine exposure. Morphine 93-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 15175841-4 2004 Subsequently, the expression of the protein product of c-fos gene (c-Fos) following naloxone administration was measured within the extended amygdala. Naloxone 84-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 15175841-4 2004 Subsequently, the expression of the protein product of c-fos gene (c-Fos) following naloxone administration was measured within the extended amygdala. Naloxone 84-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 15175841-5 2004 RESULTS: A significant increase in c-Fos immunoreactivity was seen in the centromedial amygdala (CMA), but not in the bed nucleus of the stria terminalis (BST) and the shell (AcbSh) of the nucleus accumbens (Acb) in rats treated with both morphine and naloxone. Morphine 239-247 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 15175841-5 2004 RESULTS: A significant increase in c-Fos immunoreactivity was seen in the centromedial amygdala (CMA), but not in the bed nucleus of the stria terminalis (BST) and the shell (AcbSh) of the nucleus accumbens (Acb) in rats treated with both morphine and naloxone. Naloxone 252-260 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 15334681-7 2004 RESULTS: Compared to saline, tegaserod significantly inhibited AWR in group H (0.4 mL: from 2.0 to 0.5; 0.8 mL: from 3.5 to 1.5; 1.2 mL: from 4.0 to 3.0, P<0.01), but had no significant effect on group C. Tegaserod dose-dependently attenuated the number of c-Fos positive neurons in limbic structures, anterior cingulate cortex (ACC) showed the greatest attenuation. tegaserod 29-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 260-265 15507406-3 2004 To test this hypothesis, we utilized immunohistochemical detection of nuclear c-fos expression in the myenteric and submucosal plexuses of the rat small intestine following intraintestinal infusions of oleate or glucose. Oleic Acid 202-208 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 15507406-6 2004 Oleate and glucose infusions significantly increased the number of Fos-immunoreactive nuclei in the submucosal plexus of the duodenum and jejunum, however, only glucose increased Fos-immunoreactivity in the ileum. Oleic Acid 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 15507406-6 2004 Oleate and glucose infusions significantly increased the number of Fos-immunoreactive nuclei in the submucosal plexus of the duodenum and jejunum, however, only glucose increased Fos-immunoreactivity in the ileum. Oleic Acid 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-182 15507406-6 2004 Oleate and glucose infusions significantly increased the number of Fos-immunoreactive nuclei in the submucosal plexus of the duodenum and jejunum, however, only glucose increased Fos-immunoreactivity in the ileum. Glucose 11-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 15507406-6 2004 Oleate and glucose infusions significantly increased the number of Fos-immunoreactive nuclei in the submucosal plexus of the duodenum and jejunum, however, only glucose increased Fos-immunoreactivity in the ileum. Glucose 11-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-182 15507406-6 2004 Oleate and glucose infusions significantly increased the number of Fos-immunoreactive nuclei in the submucosal plexus of the duodenum and jejunum, however, only glucose increased Fos-immunoreactivity in the ileum. Glucose 161-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-182 15507406-7 2004 Oleate and glucose infusions were associated with a small increase in Fos-immunoreactivity in NOS-immunoreactive neurons in the myenteric plexus. Oleic Acid 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 15507406-7 2004 Oleate and glucose infusions were associated with a small increase in Fos-immunoreactivity in NOS-immunoreactive neurons in the myenteric plexus. Glucose 11-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 15342266-0 2004 Administration of MK-801 decreases c-Fos expression in the trigeminal sensory nuclear complex but increases it in the midbrain during experimental movement of rat molars. Dizocilpine Maleate 18-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 15342266-2 2004 Meanwhile, MK-801, a noncompetitive antagonist of N-methyl-D-aspartate (NMDA) receptors, was shown to markedly reduce the expression of c-Fos in the trigeminal subnucleus caudalis (Vc) following noxious stimulation but to enhance c-Fos expression markedly in other brain regions, i.e., the neocortex, dorsal raphe and thalamic nuclei. Dizocilpine Maleate 11-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 15342266-2 2004 Meanwhile, MK-801, a noncompetitive antagonist of N-methyl-D-aspartate (NMDA) receptors, was shown to markedly reduce the expression of c-Fos in the trigeminal subnucleus caudalis (Vc) following noxious stimulation but to enhance c-Fos expression markedly in other brain regions, i.e., the neocortex, dorsal raphe and thalamic nuclei. Dizocilpine Maleate 11-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 230-235 15342266-3 2004 In the present study, we examined the nature of c-Fos expression in the brainstem including the TSNC and midbrain following administration of MK-801 and/or experimental movement of the rat molars. Dizocilpine Maleate 142-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 15342266-5 2004 Intraperitoneal administration of MK-801 (0.03, 0.3 and 3.0 mg/kg) prior to the onset of experimental tooth movement reduced c-Fos in the TSNC (Vc I/II, Vodm and Vor) but increased it in the nucleus raphe magnus (NRM), ventrolateral PAG (vl PAG), DR and EW. Dizocilpine Maleate 34-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 15388298-0 2004 NMDA-R1 antisense oligodeoxynucleotides modify formalin-induced nociception and spinal c-Fos expression in rat spinal cord. Oligodeoxyribonucleotides 18-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 15529010-0 2004 The AMPA receptor potentiator LY404187 increases cerebral glucose utilization and c-fos expression in the rat. LY 404187 30-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 15262987-1 2004 Membrane depolarization of skeletal muscle cells induces slow inositol trisphosphate-mediated calcium signals that regulate the activity of transcription factors such as the cAMP-response element-binding protein (CREB), jun, and fos. inositol 1,2,3-trisphosphate 62-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 229-232 15262987-1 2004 Membrane depolarization of skeletal muscle cells induces slow inositol trisphosphate-mediated calcium signals that regulate the activity of transcription factors such as the cAMP-response element-binding protein (CREB), jun, and fos. Calcium 94-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 229-232 15072955-7 2004 Cicaprost also inhibited ET-1 induction of c-fos mRNA expression, an additional marker of hypertrophy in ARCM (n = 5, P < 0.005). cicaprost 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 15336513-7 2004 CONCLUSIONS: In our animal model of schizophrenia, ketamine may evoke its stimulating effect on neurogenesis via a block of the N-methyl-D-aspartate receptor directly by reducing the c-Fos/c-Jun expression, resulting in a depression of the AP1 transcription factor complex and/or by a reduced nitric oxide production or an enhanced serotonergic activity. Ketamine 51-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 183-188 15693287-9 2004 RESULTS: The data demonstrate that in the early phase of the acute hypoglycemia, the number of the dually labeled Fos-HCRTir perikarya in the entire LHA was only moderately increased from 9.54 to 15.64% in spite of the fact that within the same period the plasma glucose levels were declined by more than 70%. Glucose 263-270 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 15305089-9 2004 In contrast to the control group, increased Fos expression was found following the use of both capsaicin and bradykinin in a variety of areas of the brain. Capsaicin 95-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 15388298-3 2004 Using antisense oligodeoxynucleotides, we tested the role of the NR1 subunit of the NMDA receptor in the nociception and expression of the immediate early gene c-fos following formalin-induced pain. Formaldehyde 176-184 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 15388298-9 2004 Similarly, the antisense oligodeoxynucleotide virtually abolished formalin-induced expression of c-Fos-like immunoreactivity (Fos-IR) in the spinal cord dorsal horn ipsilateral to injection. Oligodeoxyribonucleotides 25-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 15388298-9 2004 Similarly, the antisense oligodeoxynucleotide virtually abolished formalin-induced expression of c-Fos-like immunoreactivity (Fos-IR) in the spinal cord dorsal horn ipsilateral to injection. Formaldehyde 66-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 15388298-11 2004 We conclude that an NR1 antisense oligodeoxynucleotide inhibits both nociceptive behavior and c-fos expression following formalin injection in rats, demonstrating that NR1 plays an important role in the development of noxious stimulation induced c-fos expression in this model. Oligodeoxyribonucleotides 34-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 15388298-11 2004 We conclude that an NR1 antisense oligodeoxynucleotide inhibits both nociceptive behavior and c-fos expression following formalin injection in rats, demonstrating that NR1 plays an important role in the development of noxious stimulation induced c-fos expression in this model. Oligodeoxyribonucleotides 34-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 246-251 15388298-11 2004 We conclude that an NR1 antisense oligodeoxynucleotide inhibits both nociceptive behavior and c-fos expression following formalin injection in rats, demonstrating that NR1 plays an important role in the development of noxious stimulation induced c-fos expression in this model. Formaldehyde 121-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 15388298-11 2004 We conclude that an NR1 antisense oligodeoxynucleotide inhibits both nociceptive behavior and c-fos expression following formalin injection in rats, demonstrating that NR1 plays an important role in the development of noxious stimulation induced c-fos expression in this model. Formaldehyde 121-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 246-251 15322678-4 2004 By using immunocytochemical techniques, the formalin-induced Fos-LI neurons in the lumbar dorsal horn were calculated 1 h after formalin injection. Formaldehyde 44-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 15322678-4 2004 By using immunocytochemical techniques, the formalin-induced Fos-LI neurons in the lumbar dorsal horn were calculated 1 h after formalin injection. Formaldehyde 128-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 15322678-8 2004 Chel + Nal also exhibited a significant decrease in Fos-LI neurons in the ipsilateral dorsal horn as compared to i.t. chelerythrine 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 15322678-8 2004 Chel + Nal also exhibited a significant decrease in Fos-LI neurons in the ipsilateral dorsal horn as compared to i.t. Naloxone 7-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 15312814-8 2004 RESULTS: Withdrawal was attenuated and c-Fos, PKA, and pCREB expression was decreased in the NTS and LC of rats receiving chronic very low doses of naltrexone. Naltrexone 148-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 15529010-7 2004 LY404187 (0.5 mg/kg) also produced increases in c-fos immunoreactivity in the cortex, locus coeruleus, and the dorsal raphe nucleus. LY 404187 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 15312784-12 2004 administration of the nitric oxide (NO) inhibitor, l-NAME, indicating that endogenous NO is a necessary intermediary in CGRP and AM induced c-fos expression in the rat spinal cord. NG-Nitroarginine Methyl Ester 51-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 15306261-0 2004 Sucrose ingestion elicits reduced Fos expression in the nucleus accumbens of anhedonic rats. Sucrose 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 15142833-5 2004 Fos-positive neurons were sparse in euhydrated rats but were numerous in the SFO, MnPO, and the dorsal cap of the OVLT after 48-h water deprivation. Water 130-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 15265647-11 2004 Thus, the present results support the Fos data and indicate that the DPAG is involved in the expression of some but not all of the cardiovascular and behavioral components of the response to cat odor. dpag 69-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 15342105-5 2004 The functional implications of this pathway in cardiovascular functions were verified using Fos protein induction in response to hypotension induced by continuous intravenous administration of hydralazine-hydrochloride. Hydralazine 193-218 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 15236464-7 2004 Nitroprusside infusion significantly increased the number of Fos-IR RVLM neurons compared with saline controls. Nitroprusside 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 15236464-8 2004 In nitroprusside-treated rats, virtually all Fos/TH neurons in the RVLM were immunoreactive for CART (98% +/- 1.3%, SD; n = 7), whereas 29% +/- 8.3% of CART-positive, TH-negative neurons showed Fos immunoreactivity. Nitroprusside 3-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 15236464-8 2004 In nitroprusside-treated rats, virtually all Fos/TH neurons in the RVLM were immunoreactive for CART (98% +/- 1.3%, SD; n = 7), whereas 29% +/- 8.3% of CART-positive, TH-negative neurons showed Fos immunoreactivity. Nitroprusside 3-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 194-197 15205939-6 2004 Applications of ATP or Ca2+ ionophore A23187, both of which increase [Ca2+]i, induced immediate expression of c-fos in primary cultured chondrocytes: 1 microM ATP elicited an increase of [Ca2+]i in chondrocytes in fetal cartilage slices, but 1 mM was required in adult cartilage slices. Adenosine Triphosphate 16-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 15205939-6 2004 Applications of ATP or Ca2+ ionophore A23187, both of which increase [Ca2+]i, induced immediate expression of c-fos in primary cultured chondrocytes: 1 microM ATP elicited an increase of [Ca2+]i in chondrocytes in fetal cartilage slices, but 1 mM was required in adult cartilage slices. Calcimycin 38-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 15205939-6 2004 Applications of ATP or Ca2+ ionophore A23187, both of which increase [Ca2+]i, induced immediate expression of c-fos in primary cultured chondrocytes: 1 microM ATP elicited an increase of [Ca2+]i in chondrocytes in fetal cartilage slices, but 1 mM was required in adult cartilage slices. Adenosine Triphosphate 159-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 15305869-6 2004 The D1 antagonist, SCH23390, and the D2 antagonist, sulpiride, both attenuated expression of Fos in the medial parvocellular hypothalamic paraventricular nucleus (mpPVN) corticotropin-releasing factor cells at the apex of the HPA axis, as well as in most extra-hypothalamic brain regions examined in response to interleukin-1 beta. SCH 23390 19-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 15305869-6 2004 The D1 antagonist, SCH23390, and the D2 antagonist, sulpiride, both attenuated expression of Fos in the medial parvocellular hypothalamic paraventricular nucleus (mpPVN) corticotropin-releasing factor cells at the apex of the HPA axis, as well as in most extra-hypothalamic brain regions examined in response to interleukin-1 beta. Sulpiride 52-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 15039453-5 2004 In a rat model of Parkinson"s disease (unilateral 6-hydroxydopamine lesion), the pulmonary formulation of L-dopa (0.5-2.0 mg) yielded more rapid and robust elevations in striatal L-dopa, dopamine, and dihydroxyphenylacetic acid levels, as well as 2.5 to 3.7 times as many c-fos-expressing striatal neurons. Levodopa 106-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 272-277 15111640-0 2004 Inorganic lead exposure in the rat activates striatal cFOS expression at lower blood levels and inhibits amphetamine-induced cFOS expression at higher blood levels. Amphetamine 105-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-129 15111640-2 2004 Amphetamine (AMPH)-induced cFOS immunoreactivity (cFOS-IR) in the striatum was determined after a 3-week exposure to lead acetate (0, 50, or 250 ppm). Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-31 15111640-2 2004 Amphetamine (AMPH)-induced cFOS immunoreactivity (cFOS-IR) in the striatum was determined after a 3-week exposure to lead acetate (0, 50, or 250 ppm). Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-54 15111640-2 2004 Amphetamine (AMPH)-induced cFOS immunoreactivity (cFOS-IR) in the striatum was determined after a 3-week exposure to lead acetate (0, 50, or 250 ppm). Amphetamine 13-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-31 15111640-2 2004 Amphetamine (AMPH)-induced cFOS immunoreactivity (cFOS-IR) in the striatum was determined after a 3-week exposure to lead acetate (0, 50, or 250 ppm). Amphetamine 13-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-54 15111640-5 2004 In the untreated control (Con) group, AMPH challenge (Con/AMPH) increased cFOS-IR expression by approximately 35-fold over Veh challenge (Con/Veh) (P < 0.01). Amphetamine 38-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-78 15111640-5 2004 In the untreated control (Con) group, AMPH challenge (Con/AMPH) increased cFOS-IR expression by approximately 35-fold over Veh challenge (Con/Veh) (P < 0.01). Amphetamine 58-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-78 15111640-8 2004 The increase in cFOS-IR in the Pb50/AMPH was also significant (P < 0.01), but it was not different from the Con/AMPH (P > 0.20). Amphetamine 36-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-20 15111640-11 2004 However, chronic 250 ppm lead exposure inhibited AMPH-induced activation of cFOS in the striatum by about 89%. Amphetamine 49-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-80 15111640-12 2004 Therefore, lead is capable of both activating cFOS expression at low levels of exposure (mean blood lead level 21.6 +/- 1.9 microg/dl) and inhibiting AMPH-induced cFOS expression at higher levels of exposure (mean blood lead level 47.4 +/- 2.6 microg/dl). Amphetamine 150-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-167 15288415-0 2004 c-fos and CRF receptor gene transcription in the brain of acetic acid-induced somato-visceral pain in rats. Acetic Acid 58-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 15259085-0 2004 Expression and localization of c-Fos and NOS in the central nerve system following esophageal acid stimulation in rats. esophageal acid 83-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 15059790-0 2004 Prolonged retention of the anorectic cobalt protoporphyrin in the hypothalamus and the resulting expression of Fos. cobaltiprotoporphyrin 37-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 15059790-2 2004 The goal of this work was to determine 1) if the prolonged duration of action of CoPP is related to its prolonged retention within the brain; and 2) with the use of immunohistochemical detection of Fos, the product of the early-immediate gene c-fos, which cells are activated after exposure to CoPP. cobaltiprotoporphyrin 81-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 198-201 15059790-2 2004 The goal of this work was to determine 1) if the prolonged duration of action of CoPP is related to its prolonged retention within the brain; and 2) with the use of immunohistochemical detection of Fos, the product of the early-immediate gene c-fos, which cells are activated after exposure to CoPP. cobaltiprotoporphyrin 81-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 243-248 15259085-1 2004 AIM: To determine the distribution of neurons expressing c-Fos and nitric oxide synthase (NOS) in the central nerve system (CNS) following esophageal acid exposure, and to investigate the relationship between c-Fos and NOS. esophageal acid 139-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 15223361-4 2004 We also studied the effect of CeA-injected DAMGO on LiCl-induced increases in c-Fos IR in the amygdala. Lithium Chloride 52-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 15223361-8 2004 We also found that intra-CeA injection of DAMGO, prior to LiCl injection, decreased c-Fos IR levels in the CeA compared to vehicle-injected rats. Enkephalin, Ala(2)-MePhe(4)-Gly(5)- 42-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 15232294-2 2004 Either the PAR-2 agonist SLIGRL-NH2 or capsaicin, injected into the parotid duct, caused expression of Fos in the trigeminal subnucleus caudalis, although the PAR-2-inactive reversed peptide had no such effect. seryl-leucyl-isoleucyl-glycyl--arginyl-leucinamide 25-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-106 15232294-2 2004 Either the PAR-2 agonist SLIGRL-NH2 or capsaicin, injected into the parotid duct, caused expression of Fos in the trigeminal subnucleus caudalis, although the PAR-2-inactive reversed peptide had no such effect. Capsaicin 39-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-106 15232294-3 2004 The Fos expression caused by PAR-2 activation was inhibited by ablation of capsaicin-sensitive sensory neurons. Capsaicin 75-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-7 15193999-7 2004 The effect of FK960 on c-Fos was inhibited by PD98059 (10microM), an ERK kinase inhibitor, and cycloheximide (1microg/ml), a transcription inhibitor, and the effect of FK960 on CREB phosphorylation was blocked by PD98059. N-(4-acetyl-1-piperazinyl) -4-fluorobenzamide monohydrate 14-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 15193999-7 2004 The effect of FK960 on c-Fos was inhibited by PD98059 (10microM), an ERK kinase inhibitor, and cycloheximide (1microg/ml), a transcription inhibitor, and the effect of FK960 on CREB phosphorylation was blocked by PD98059. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 46-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 15193999-7 2004 The effect of FK960 on c-Fos was inhibited by PD98059 (10microM), an ERK kinase inhibitor, and cycloheximide (1microg/ml), a transcription inhibitor, and the effect of FK960 on CREB phosphorylation was blocked by PD98059. Cycloheximide 95-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 15193999-9 2004 These results suggest that FK960 stimulates GDNF production in c-Fos- and CREB-dependent mechanisms in cultured astrocytes and that ERK signal is responsible for both c-Fos expression and CREB phosphorylation in the cascades. N-(4-acetyl-1-piperazinyl) -4-fluorobenzamide monohydrate 27-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 14684379-5 2004 The c-fos expression in all these brain nuclei was blocked by truncal vagotomy as well as by perivagal capsaicin treatment, suggesting that vagal afferent pathways may mediate this response. Capsaicin 103-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 14684379-6 2004 Intravenous injection of 5-HT3 receptor antagonist granisetron blocked c-fos expression in all brain nuclei examined, although intracerebroventricular injection of granisetron had no effect, suggesting that 5-HT released from the stomach may activate 5-HT3 receptors located in the peripheral vagal afferent nerve terminals and then induce brain c-fos expression. Granisetron 51-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 14684379-6 2004 Intravenous injection of 5-HT3 receptor antagonist granisetron blocked c-fos expression in all brain nuclei examined, although intracerebroventricular injection of granisetron had no effect, suggesting that 5-HT released from the stomach may activate 5-HT3 receptors located in the peripheral vagal afferent nerve terminals and then induce brain c-fos expression. Granisetron 51-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 346-351 15281522-0 2004 Intraarticular pretreatment with ketamine and memantine could prevent arthritic pain: relevance to the decrease of spinal c-fos expression in rats. Ketamine 33-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 15259085-10 2004 c-Fos and NOS had significant correlation between PVN, PBN, NTS/DMV, RNM and AP. (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 64-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 15249992-0 2004 Changes in c-fos expression in the rat heart during morphine withdrawal. Morphine 52-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 15249992-2 2004 We previously demonstrated an increase in Fos expression in the heart during morphine withdrawal. Morphine 77-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 15249992-3 2004 In the present study we examined the role of beta- and alpha-adrenoceptors in naloxone-precipitated increases in Fos expression in the heart. Naloxone 78-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-116 15249992-6 2004 Using immunohistochemical staining of Fos, the present results indicate that morphine withdrawal induced marked Fos immunoreactivity (Fos-IR) within the cardiomyocyte nuclei. Morphine 77-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 15249992-6 2004 Using immunohistochemical staining of Fos, the present results indicate that morphine withdrawal induced marked Fos immunoreactivity (Fos-IR) within the cardiomyocyte nuclei. Morphine 77-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 15249992-6 2004 Using immunohistochemical staining of Fos, the present results indicate that morphine withdrawal induced marked Fos immunoreactivity (Fos-IR) within the cardiomyocyte nuclei. Morphine 77-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 15249992-7 2004 Moreover, Western blot analysis revealed a peak expression of c-fos in the right and left ventricles after naloxone-precipitated withdrawal in parallel with an increase in noradrenaline (NA) turnover. Naloxone 107-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 15249992-7 2004 Moreover, Western blot analysis revealed a peak expression of c-fos in the right and left ventricles after naloxone-precipitated withdrawal in parallel with an increase in noradrenaline (NA) turnover. Norepinephrine 172-185 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 15249992-10 2004 However, pre-treatment with alpha2-adrenoceptor antagonist, yohimbine (1 mg/kg intraperitoneally), 20 min before naloxone administration to morphine-dependent rats antagonized Fos expression and the enhancement of NA turnover in the heart. Yohimbine 60-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 176-179 15158372-0 2004 Cannabidiol increases Fos expression in the nucleus accumbens but not in the dorsal striatum. Cannabidiol 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-25 15158372-2 2004 In the present paper we employed the detection of Fos protein to investigate neuronal activation in the dorsal striatum and nucleus accumbens of male Wistar rats after systemic administration of CBD (120 mg/kg), haloperidol (1 mg/kg) or clozapine (20 mg/kg). Cannabidiol 195-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 15158372-2 2004 In the present paper we employed the detection of Fos protein to investigate neuronal activation in the dorsal striatum and nucleus accumbens of male Wistar rats after systemic administration of CBD (120 mg/kg), haloperidol (1 mg/kg) or clozapine (20 mg/kg). Haloperidol 212-223 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 15158372-2 2004 In the present paper we employed the detection of Fos protein to investigate neuronal activation in the dorsal striatum and nucleus accumbens of male Wistar rats after systemic administration of CBD (120 mg/kg), haloperidol (1 mg/kg) or clozapine (20 mg/kg). Clozapine 237-246 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 15158372-3 2004 Only haloperidol was able to increase the number of Fos immunoreactive neurons (FIr) in the dorsal striatum (vehicle: 0.07 +/- 0.07/0.1 mm(2), haloperidol: 28.3 +/- 8.9/0.1 mm(2), p < 0.01). Haloperidol 5-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 15107478-9 2004 Superoxide dismutase mimetic, a potent scavenger of superoxide anions, prevented IH-induced c-fos, AP-1 and TH activations. Superoxides 52-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 15107478-12 2004 Pharmacological inhibitors of complex I mimicked the effects of IH during normoxia and occluded the effects of IH on c-fos activation, suggesting the involvement of the mitochondrial electron transport chain in the generation of superoxide anions during IH. Superoxides 229-246 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 15188517-7 2004 Intra-gastric administration of tegaserod (2 mg/kg.d) resulted in a significant decrease of Fos labeled neurons (22.0+/-7.7) and SP density (12.5+/-1.4) in the dorsal horn in the lumbarsacral spinal cord compared to those of the control group (62.2+/-18.9, 35.9+/-8.9, P<0.05). tegaserod 32-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 15140652-0 2004 Single footshock attenuates c-Fos expression induced by 5-HT2A receptor agonist (+/-)-2,5-dimethoxy-4-iodoamphetamine hydrochloride in rat brain. 4-iodo-2,5-dimethoxyphenylisopropylamine 80-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 15140652-1 2004 Administration of 3 mg/kg 5-hydroxytryptamine2A/2C (5-HT2A/2C) receptor agonist (+/-)-2,5-dimethoxy-4-iodoamphetamine hydrochloride (DOI) induced c-Fos expression in all areas of the rat neocortex. 4-iodo-2,5-dimethoxyphenylisopropylamine 80-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-151 15240353-5 2004 DOI and 8-OH-DPAT also produced a marked induction of c-Fos in the paraventricular nucleus (PVN), but the induction was not different if the two compounds were given together. 8-Hydroxy-2-(di-n-propylamino)tetralin 8-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 15136051-0 2004 Inhibitory effects of tetrandrine on the serum- and platelet-derived growth factor-BB-induced proliferation of rat aortic smooth muscle cells through inhibition of cell cycle progression, DNA synthesis, ERK1/2 activation and c-fos expression. tetrandrine 22-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 225-230 15136051-6 2004 In accordance with these findings, TET 5 microM caused a 48% decrease in the early elevation of c-fos expression induced after 10% FBS addition. tetrandrine 35-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 14978197-8 2004 Finally, the pretreatment of VSMCs with PTX or cilostamide, but not denbufylline, reduced TNF-alpha-induced CCL2/MCP-1 production, which was preceded by attenuation of p65/NF-kappaB nuclear translocation, p42/44 MAPK, and JNK-c-Jun phosphorylation, and c-Fos up-regulation. vsmcs 29-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 253-258 14978197-8 2004 Finally, the pretreatment of VSMCs with PTX or cilostamide, but not denbufylline, reduced TNF-alpha-induced CCL2/MCP-1 production, which was preceded by attenuation of p65/NF-kappaB nuclear translocation, p42/44 MAPK, and JNK-c-Jun phosphorylation, and c-Fos up-regulation. Pentoxifylline 40-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 253-258 14978197-8 2004 Finally, the pretreatment of VSMCs with PTX or cilostamide, but not denbufylline, reduced TNF-alpha-induced CCL2/MCP-1 production, which was preceded by attenuation of p65/NF-kappaB nuclear translocation, p42/44 MAPK, and JNK-c-Jun phosphorylation, and c-Fos up-regulation. cilostamide 47-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 253-258 15275954-7 2004 In infralimbic cortex, rats, which learned the reward value of the olfactory cue and were water-reinforced the day of sacrifice, showed a higher Fos expression. Water 90-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-148 15217452-0 2004 Intravesical resiniferatoxin decreases spinal c-fos expression and increases bladder volume to reflex micturition in rats with chronic inflamed urinary bladders. resiniferatoxin 13-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 15217452-1 2004 OBJECTIVE: To evaluate the effect of intravesical resiniferatoxin on spinal c-fos expression and bladder volume at reflex micturition in rats with chronic urinary bladder inflammation. resiniferatoxin 50-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 15217452-8 2004 CONCLUSION: Intravesical resiniferatoxin decreases c-fos expression and increases bladder capacity in chronically inflamed rat bladders. resiniferatoxin 25-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 15240549-10 2004 Antisense RIalpha in the liver induced the phase II enzymes, glutathione S-transferase and quinone oxidoreductase, c-fos protein, and activator protein 1 (AP-1)- and cAMP response element (CRE)-directed transcription. rialpha 10-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 15233757-6 2004 Intrathecal and intravenous injection of ZJ-11 suppressed the expression of Fos-like immunoreactivity, induced by paw formalin injection, in laminae I-II in segments L4-L5 of the spinal cord, suggesting an action on sensory spinal transmission. zj-11 41-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 15233757-6 2004 Intrathecal and intravenous injection of ZJ-11 suppressed the expression of Fos-like immunoreactivity, induced by paw formalin injection, in laminae I-II in segments L4-L5 of the spinal cord, suggesting an action on sensory spinal transmission. Formaldehyde 118-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 15153782-10 2004 In addition, nicotinamide diminished c-fos and zif268 immediate-early gene expressions following OGD. Niacinamide 13-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 14973310-8 2004 Furthermore, as predicted by the isodendritic morphology of cells in the ventromedial medulla, midline bicuculline microinjection increased the number of c-fos immunoreactive cells in both midline raphe and lateral reticular nuclei. midline 95-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 14973310-8 2004 Furthermore, as predicted by the isodendritic morphology of cells in the ventromedial medulla, midline bicuculline microinjection increased the number of c-fos immunoreactive cells in both midline raphe and lateral reticular nuclei. Bicuculline 103-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 14764439-5 2004 After 1 h of stimulation with PE, mRNA expression of c-Fos and c-Jun was upregulated to 185 +/- 32 and 132 +/- 13% of control. Phenylephrine 30-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 15281522-0 2004 Intraarticular pretreatment with ketamine and memantine could prevent arthritic pain: relevance to the decrease of spinal c-fos expression in rats. Memantine 46-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 15281522-3 2004 The intraarticular injection of ketamine (1 mg) or memantine (0.2 mg) also suppressed c-Fos expression in the laminae I-II and laminae V-VI at the L3-4 spinal level. Ketamine 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 15281522-3 2004 The intraarticular injection of ketamine (1 mg) or memantine (0.2 mg) also suppressed c-Fos expression in the laminae I-II and laminae V-VI at the L3-4 spinal level. Memantine 51-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 15155837-6 2004 In cultured smooth muscle cells from rat aorta, forskolin induced a rapid increase in Fos/p-Fos protein levels and activator protein 1 (AP-1) binding activity. Colforsin 48-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 15181236-4 2004 As for the NGF-induced neurite outgrowth, the CREB-cAMP responsive element (CRE) pathway is important to the activation of immediate-early genes such as c-fos. Cyclic AMP 51-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-158 15330290-0 2004 Dizocilpine blocks the effects of delta sleep-inducing peptide-induced suppression of c-fos gene expression in the paraventricular nucleus of the hypothalamus in rats. Dizocilpine Maleate 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 15330290-1 2004 The characteristics of the actions of the non-competitive blocker of NMDA receptors dizocilpine on the expression of the early c-fos gene in the paraventricular nuclei of the hypothalamus were studied in rats with different predicted susceptibilities to emotional stress in conditions of treatment with delta sleep-inducing peptide. Dizocilpine Maleate 84-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 15330290-2 2004 The results showed that prior treatment with dizocilpine blocked reductions in c-fos expression induced by delta sleep peptide. Dizocilpine Maleate 45-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 15155837-6 2004 In cultured smooth muscle cells from rat aorta, forskolin induced a rapid increase in Fos/p-Fos protein levels and activator protein 1 (AP-1) binding activity. Colforsin 48-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 15155837-8 2004 We conclude that forskolin/cAMP decrease the expression of heterodimeric sGC in rat aortic smooth muscle cells via activation of Fos/AP-1, which decreases the expression of HuR and thus destabilizes the sGCalpha1 and beta1 mRNA. Colforsin 17-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-132 15155837-8 2004 We conclude that forskolin/cAMP decrease the expression of heterodimeric sGC in rat aortic smooth muscle cells via activation of Fos/AP-1, which decreases the expression of HuR and thus destabilizes the sGCalpha1 and beta1 mRNA. Cyclic AMP 27-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-132 15706927-0 2004 [The influence of CGE on expression of IL-1beta and c-fos protein in the hippocampus region in rats following cerebral ischemia-reperfusion]. Clopidogrel 18-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 15706927-1 2004 OBJECTIVE: To observe inhibiting effect of CGE (compound ginseng extract) on increased expression of IL-1beta and c-fos protein following cerebral ischemia-reperfusion. Clopidogrel 43-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 15706927-13 2004 The effect of CGE is associated with its dosage, i.e. a larger dosage has a better effect on expression of c-fos protein in post-stroke dementia. Clopidogrel 14-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 15129159-0 2004 FOS expression in orexin neurons following muscimol perfusion of preoptic area. Muscimol 43-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 15120589-5 2004 Our results showed that the substantial increase in c-fos expression in the shell and core induced by haloperidol treatment was detected by both stereological and profile counting methods; in contrast, the weaker effect of clozapine on c-fos expression was detected differentially by the two methods. Haloperidol 102-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 15120589-5 2004 Our results showed that the substantial increase in c-fos expression in the shell and core induced by haloperidol treatment was detected by both stereological and profile counting methods; in contrast, the weaker effect of clozapine on c-fos expression was detected differentially by the two methods. Clozapine 223-232 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 236-241 15120589-6 2004 Whereas the profile counting method reported a reduction of c-fos in the core by clozapine, and an increase in c-fos in the lateral septum, these effects were not replicated using stereology. Clozapine 81-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 15147776-0 2004 Effect of naloxone-precipitated morphine withdrawal on c-fos expression in rat corticotropin-releasing hormone neurons in the paraventricular hypothalamus and extended amygdala. Naloxone 10-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 14978197-5 2004 PD98059 abolished TNF-alpha-activated p42/44 MAPK phosphorylation and c-Fos up-regulation, whereas SP600125 inhibited TNF-alpha-activated JNK and c-Jun phosphorylation. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 15147776-0 2004 Effect of naloxone-precipitated morphine withdrawal on c-fos expression in rat corticotropin-releasing hormone neurons in the paraventricular hypothalamus and extended amygdala. Morphine 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 15147776-3 2004 Here, by means of dual-immunohistochemical methodology, we examined the co-expression of the c-Fos protein and CRH following naloxone-precipitated morphine withdrawal. Naloxone 125-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 15086447-4 2004 RESULTS: Stimulation with fetal bovine serum (FBS), phorbol 12-myristate 13-acetate (PMA) and ET-1 caused about a doubling of c-fos mRNA. Tetradecanoylphorbol Acetate 52-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 15147776-3 2004 Here, by means of dual-immunohistochemical methodology, we examined the co-expression of the c-Fos protein and CRH following naloxone-precipitated morphine withdrawal. Morphine 147-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 15147776-6 2004 We found that naloxone-precipitated withdrawal of morphine-dependent rats increased c-Fos immunoreactivity (IR) in CRH positive neurons in the paraventricular hypothalamus. Naloxone 14-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 15147776-6 2004 We found that naloxone-precipitated withdrawal of morphine-dependent rats increased c-Fos immunoreactivity (IR) in CRH positive neurons in the paraventricular hypothalamus. Morphine 50-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 15147776-7 2004 Withdrawal of morphine-dependent rats also increased c-Fos-IR in the central amygdala and bed nucleus of the stria terminalis, however these were in CRH negative neurons. Morphine 14-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 15086447-4 2004 RESULTS: Stimulation with fetal bovine serum (FBS), phorbol 12-myristate 13-acetate (PMA) and ET-1 caused about a doubling of c-fos mRNA. Tetradecanoylphorbol Acetate 85-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 15086447-5 2004 The ET-1-induced c-fos expression was steady (15-60 min) and was inhibited by the inhibitor of the ET(A) receptor, BQ-123. cyclo(Trp-Asp-Pro-Val-Leu) 115-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 15086447-10 2004 In contrast, ET-1-induced c-fos expression was inhibited by the calcium chelator BAPTA, suggesting a role for intracellular calcium. Calcium 64-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 15086447-10 2004 In contrast, ET-1-induced c-fos expression was inhibited by the calcium chelator BAPTA, suggesting a role for intracellular calcium. 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid 81-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 15086447-10 2004 In contrast, ET-1-induced c-fos expression was inhibited by the calcium chelator BAPTA, suggesting a role for intracellular calcium. Calcium 124-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 15113957-1 2004 In the present study, we tested the hypothesis that dietary oligofructose (FOS) can modulate both the response to an endotoxic shock induced by lipopolysaccharide (LPS) administration and the activity of resident hepatic macrophages, i.e., Kupffer cells. oligofructose 60-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 15113957-5 2004 After LPS injection, serum levels of tumor necrosis factor (TNF)-alpha, a proinflammatory cytokine, and prostaglandin E(2) (PGE(2)), an immunosuppressive mediator, were higher in FOS-treated rats than in control rats. Dinoprostone 104-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-182 15113957-7 2004 FOS treatment increased the number of large phagocytic Kupffer cells, as assessed by histological examination of the liver after colloidal carbon injection into the portal vein. Carbon 139-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 15113957-8 2004 Precision-cut liver slices (PCLS) from FOS-treated rats released more TNF-alpha and PGE(2) into the incubation medium than PCLS from control rats, independently of LPS challenge in vitro. Prostaglandins E 84-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 15117340-0 2004 Acute intracarotid glucose injection towards the brain induces specific c-fos activation in hypothalamic nuclei: involvement of astrocytes in cerebral glucose-sensing in rats. Glucose 19-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 15279077-4 2004 METHODS: c-Fos-ir was analyzed by immunohistochemical methods in formaline-fixed, paraffin-embedded sections of rat pancreatic beta cells (BCs), in adrenal medullary chromaffin cells (CCs) that are derived from neural crest, in exocrine pancreatic acinar cells (ACs) that are derived from endoderm, and in adrenal cortex zona reticularis cells (RCs) that are derived from mesoderm. Formaldehyde 65-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 15279077-4 2004 METHODS: c-Fos-ir was analyzed by immunohistochemical methods in formaline-fixed, paraffin-embedded sections of rat pancreatic beta cells (BCs), in adrenal medullary chromaffin cells (CCs) that are derived from neural crest, in exocrine pancreatic acinar cells (ACs) that are derived from endoderm, and in adrenal cortex zona reticularis cells (RCs) that are derived from mesoderm. Paraffin 82-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 15279077-4 2004 METHODS: c-Fos-ir was analyzed by immunohistochemical methods in formaline-fixed, paraffin-embedded sections of rat pancreatic beta cells (BCs), in adrenal medullary chromaffin cells (CCs) that are derived from neural crest, in exocrine pancreatic acinar cells (ACs) that are derived from endoderm, and in adrenal cortex zona reticularis cells (RCs) that are derived from mesoderm. chromaffin 166-176 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 15341219-0 2004 Expression of the c-fos gene in spinal cord and brain cells in rats subjected to stress in conditions of exposure to various types of halothane anesthesia. Halothane 134-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 15341219-2 2004 Synthesis of c-Fos-like proteins occurred only in the spinal cord in conditions of constant 1.5% halothane anesthesia. Halothane 97-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 15341219-3 2004 Use of induction anesthesia with 1.5% halothane allowed detection of c-Fos-like protein expression in cells of the rat spinal cord (lumbar segments) and brain, both when animals were placed in a hammock and when mechanical pain stimulation or electromagnetic irradiation of the skin with UHF currents were applied. Halothane 38-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 15115808-7 2004 Midazolam inhibited Fos expression in key limbic regions involved in pheromone transduction (medial amygdala and bed nucleus of the stria terminalis) and defensive behavior (prelimbic cortex, lateral septum, lateral and medial preoptic areas, and dorsal premammillary nucleus). Midazolam 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 15033432-1 2004 To test the hypothesis that the hippocampus field CA1 is recruited in nociceptive intensity-dependent fashion in the formalin model of inflammatory pain, we determined the effect of injection of formalin (0.625-2.5%) on the induction of Fos protein along the length of the hippocampus. Formaldehyde 195-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 237-240 15158023-0 2004 The KCNQ channel activator retigabine blocks haloperidol-induced c-Fos expression in the striatum of the rat. ezogabine 27-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 15158023-0 2004 The KCNQ channel activator retigabine blocks haloperidol-induced c-Fos expression in the striatum of the rat. Haloperidol 45-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 15158023-3 2004 This study was carried out to determine whether retigabine inhibited haloperidol-dependent activation of neurons in the striatum as measured by expression of c-Fos. ezogabine 48-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 15158023-7 2004 Haloperidol produced a large increase in the number of c-Fos-positive nuclei in different degrees in all parts of the striatum. Haloperidol 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 15158023-8 2004 Pretreatment with retigabine completely blocked haloperidol-induced c-Fos in both the ventral and dorsal striatum suggesting that retigabine via activation of the KCNQ channel interacts with haloperidol and inhibits neuronal excitation in the striatum. ezogabine 18-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 15158023-8 2004 Pretreatment with retigabine completely blocked haloperidol-induced c-Fos in both the ventral and dorsal striatum suggesting that retigabine via activation of the KCNQ channel interacts with haloperidol and inhibits neuronal excitation in the striatum. Haloperidol 48-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 15158023-8 2004 Pretreatment with retigabine completely blocked haloperidol-induced c-Fos in both the ventral and dorsal striatum suggesting that retigabine via activation of the KCNQ channel interacts with haloperidol and inhibits neuronal excitation in the striatum. ezogabine 130-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 15158023-8 2004 Pretreatment with retigabine completely blocked haloperidol-induced c-Fos in both the ventral and dorsal striatum suggesting that retigabine via activation of the KCNQ channel interacts with haloperidol and inhibits neuronal excitation in the striatum. Haloperidol 191-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 15063162-4 2004 The cortical c-Fos/ICER expression induced by lithium was not modulated by L-NAME pretreatment. Lithium 46-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 15063162-5 2004 However, lithium-induced medullary expression of c-Fos was attenuated by central L-NAME, and ICER by systemic L-NAME. Lithium 9-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 15063162-5 2004 However, lithium-induced medullary expression of c-Fos was attenuated by central L-NAME, and ICER by systemic L-NAME. NG-Nitroarginine Methyl Ester 81-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 15063162-5 2004 However, lithium-induced medullary expression of c-Fos was attenuated by central L-NAME, and ICER by systemic L-NAME. NG-Nitroarginine Methyl Ester 110-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 15063162-6 2004 These results suggest that nitric oxide is, at least partly, involved in lithium-induced c-Fos/ICER expression in the adrenal medulla, and that central nitric oxide may play a different role from peripheral nitric oxide in lithium-induced activation of adrenal medulla. Nitric Oxide 27-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 15063162-6 2004 These results suggest that nitric oxide is, at least partly, involved in lithium-induced c-Fos/ICER expression in the adrenal medulla, and that central nitric oxide may play a different role from peripheral nitric oxide in lithium-induced activation of adrenal medulla. Lithium 73-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 15063162-1 2004 This study was conducted to determine if nitric oxide (NO) is involved in lithium-induced expression of c-Fos and inducible cAMP early repressor (ICER) in the adrenal gland. Nitric Oxide 41-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 15063162-1 2004 This study was conducted to determine if nitric oxide (NO) is involved in lithium-induced expression of c-Fos and inducible cAMP early repressor (ICER) in the adrenal gland. Lithium 74-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 15033432-2 2004 Compared to injection of saline, injection of formalin (0.625-2.5%) evoked a concentration-dependent increase in nociceptive behavior and a significant linear increase in the number of Fos-positive cells in the spinal cord, especially in the deeper laminae. Formaldehyde 46-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 185-188 15033432-3 2004 Injection of saline also increased induction of Fos along the length of hippocampus. Sodium Chloride 13-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 15033432-4 2004 On the other hand, injection of formalin decreased the number of Fos-positive cells in whole CA1, CA3 and dentate gyrus, with a greater significant effect in the posterior-ventral regions of the hippocampus. Formaldehyde 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 15033432-7 2004 Of the regions examined, only the formalin-induced changes in Fos cell counts in the posterior and ventral CA1 were tightly correlated with the changes observed in the spinal cord. Formaldehyde 34-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 14668212-3 2004 In the first study, we established that sequential estradiol and progesterone treatment induces a proestrous-like rise in LH secretion and in the percentage of GnRH neurons that express Fos in grass rats, as is the case in lab rats. Estradiol 51-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 186-189 15041605-0 2004 The effects of pretreatment with lidocaine or bupivacaine on the spatial and temporal expression of c-Fos protein in the spinal cord caused by plantar incision in the rat. Lidocaine 33-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-113 15041605-0 2004 The effects of pretreatment with lidocaine or bupivacaine on the spatial and temporal expression of c-Fos protein in the spinal cord caused by plantar incision in the rat. Bupivacaine 46-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-113 15041605-5 2004 In the saline group, Fos expression was detected predominantly in laminae I-II and V-VI, and the total Fos expression was maximal at 1 h and then decreased gradually. Sodium Chloride 7-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 15041605-5 2004 In the saline group, Fos expression was detected predominantly in laminae I-II and V-VI, and the total Fos expression was maximal at 1 h and then decreased gradually. Sodium Chloride 7-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-106 14668212-3 2004 In the first study, we established that sequential estradiol and progesterone treatment induces a proestrous-like rise in LH secretion and in the percentage of GnRH neurons that express Fos in grass rats, as is the case in lab rats. Progesterone 65-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 186-189 14656894-6 2004 Angiotensin II (Ang II) stimulation increased c-Fos, c-Jun, and Sp1 binding to the MRE by 100-, 4.9-, and 1.9-fold, respectively, and these responses were inhibited by PD98059 and AT1 receptor antagonist candesartan. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 168-175 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 15068814-7 2004 The vitamin E deficiency-mediated loss of AP-1 activity was not due to an alteration in the dimeric composition of constituent proteins, but rather to a general down-regulation of steady-state levels of members of the Fos and Jun families of proteins. Vitamin E 4-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 218-221 15100610-0 2004 Ethanol-induced Fos immunoreactivity in the extended amygdala and hypothalamus of the rat brain: focus on cholinergic interneurons of the nucleus accumbens. Ethanol 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 15100610-1 2004 BACKGROUND: The primary goal of this study was to investigate the effects of varying doses of ethanol on cellular activation, as measured by Fos immunoreactivity, in brain areas that have been implicated in the reinforcing and anxiolytic effects of substance abuse and dependence, namely, the extended amygdala and hypothalamus. Ethanol 94-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 15100610-8 2004 RESULTS: A dose of 2 g/kg of ethanol significantly increased the number of Fos-immunoreactive neurons in the central nucleus of the amygdala by 149%, in the shell nucleus accumbens by 80%, and in the paraventricular nucleus of the hypothalamus by 321%. Ethanol 29-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 15100610-9 2004 Additionally, 1 g/kg of ethanol significantly increased the percentage of Fos-immunoreactive cholinergic neurons in the nucleus accumbens by 59%. Ethanol 24-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 15056282-0 2004 The protein phosphatase 1/2A inhibitor okadaic acid increases CREB and Elk-1 phosphorylation and c-fos expression in the rat striatum in vivo. Okadaic Acid 39-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 15056282-3 2004 In this study, using semi-quantitative immunohistochemical and western blot analyses and in situ hybridization histochemistry, we found that intrastriatal infusion of the protein phosphatase 1/2A inhibitor okadaic acid (0.005, 0.05 and 0.5 nmol) increased CREB and Elk-1 phosphorylation and c-Fos immunoreactivity in the injected dorsal striatum in a dose-dependent manner. Okadaic Acid 206-218 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 291-296 15056282-4 2004 In addition, okadaic acid (0.05 and 0.5 nM) increased c-fos mRNA expression in the dorsal striatum in a dose-dependent manner. Okadaic Acid 13-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 15056473-4 2004 In the present study, the modulation of D(2) receptors in the graft by host-derived glutamatergic afferents via NMDA receptors was investigated using haloperidol-induced c-Fos expression. Haloperidol 150-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-175 15056263-3 2004 RESULTS: Cyclosporin exposure increases c-fos and c-jun mRNA in the rat kidney but not in the liver. Cyclosporine 9-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 15056263-4 2004 Furthermore, chronic CsA exposure causes a further increase in c-fos and c-jun mRNA and increases the renal expression of transforming growth factor-beta (TGF-beta) mRNA. Cyclosporine 21-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 15056263-6 2004 The combined insult of ischaemia and CsA resulted in synergistic increases in c-fos, suggesting that CsA recruited a pathway for c-fos activation different from ischaemia. Cyclosporine 37-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 15056263-6 2004 The combined insult of ischaemia and CsA resulted in synergistic increases in c-fos, suggesting that CsA recruited a pathway for c-fos activation different from ischaemia. Cyclosporine 37-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 15056473-5 2004 The results indicate that haloperidol induces c-Fos in a large number of neurons in the P-zones of the graft and this induction is significantly suppressed by pretreatment with the NMDA receptor antagonist, MK-801. Haloperidol 26-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 15056263-6 2004 The combined insult of ischaemia and CsA resulted in synergistic increases in c-fos, suggesting that CsA recruited a pathway for c-fos activation different from ischaemia. Cyclosporine 101-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 15056473-5 2004 The results indicate that haloperidol induces c-Fos in a large number of neurons in the P-zones of the graft and this induction is significantly suppressed by pretreatment with the NMDA receptor antagonist, MK-801. Dizocilpine Maleate 207-213 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 15056263-6 2004 The combined insult of ischaemia and CsA resulted in synergistic increases in c-fos, suggesting that CsA recruited a pathway for c-fos activation different from ischaemia. Cyclosporine 101-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 15056263-7 2004 The calcium channel blocker, verapamil, blocked CsA-induced expression of c-fos and c-jun mRNA, and reduced the amount of TGF-beta expression. Verapamil 29-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 15066018-1 2004 Challenge of the rat gastric mucosa with HCl stimulates intrinsic neurones in the myenteric plexus of the stomach as demonstrated by immunohistochemical detection of c-Fos. Hydrochloric Acid 41-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-171 15056263-7 2004 The calcium channel blocker, verapamil, blocked CsA-induced expression of c-fos and c-jun mRNA, and reduced the amount of TGF-beta expression. Cyclosporine 48-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 15066018-3 2004 Intragastric (IG) administration of 0.5 m HCl caused c-Fos expression in 12% of myenteric neurones, whereas IG saline failed to induce c-Fos. Hydrochloric Acid 42-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 15066020-3 2004 Peripheral injection of CRF or urocortin inhibits gastric emptying and motility through interaction with CRF2 receptors and stimulates colonic transit, motility, Fos expression in myenteric neurones and defecation through activation of CRF1 receptors. Urocortins 31-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-165 15066018-6 2004 Pretreatment with capsaicin or hexamethonium, combination of both pretreatments or vagotomy reduced HCl-induced c-Fos expression by 54%, 66%, 63% and 68%, respectively. Capsaicin 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 15066018-6 2004 Pretreatment with capsaicin or hexamethonium, combination of both pretreatments or vagotomy reduced HCl-induced c-Fos expression by 54%, 66%, 63% and 68%, respectively. Hexamethonium 31-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 15066018-6 2004 Pretreatment with capsaicin or hexamethonium, combination of both pretreatments or vagotomy reduced HCl-induced c-Fos expression by 54%, 66%, 63% and 68%, respectively. Hydrochloric Acid 100-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 15010207-7 2004 reduced AMPH-stimulated c-fos mRNA levels in the dorsal (caudoputamen) and ventral (nucleus accumbens) striatum as revealed by quantitative in situ hybridization. Amphetamine 8-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 15303649-0 2004 [Effect of ecdysterone on the expression of c-fos in the brain of rats induced by microinjection beta-AP25-35 into the hippocampus]. beta-ap25-35 97-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 15010207-11 2004 These results indicate that an mGluR5-dependent mechanism selectively contributes to c-fos expression in the striatum and cortex in response to acute exposure to AMPH. Amphetamine 162-166 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 15303649-1 2004 AIM: To observe the behavior in learning and memory and the expression of c-fos gene from the brain of rats induced by beta-AP25-35, and the intervention of ecdysterone, in order to explore the protective mechanism of ecdysterone on the dysfunction of learning and memory of the rat induced by beta-AP25-35. beta-ap25 119-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 15303649-1 2004 AIM: To observe the behavior in learning and memory and the expression of c-fos gene from the brain of rats induced by beta-AP25-35, and the intervention of ecdysterone, in order to explore the protective mechanism of ecdysterone on the dysfunction of learning and memory of the rat induced by beta-AP25-35. beta-ap25 294-303 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 14972655-2 2004 In cycling rats, rising endogenous E2 levels not only induce a surge in LH release, but also increase the expression of the immediate early gene Fos in the anteroventral preoptic area (AVPV) and within gonadotropin releasing hormone (GnRH) neurons. Estradiol 35-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-148 15303649-8 2004 Ecdysterone was shown to improve the learning and memory of the rats and increase the expression of c-fos. Ecdysterone 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 15026155-4 2004 We examined the effect of acupuncture on nicotine-induced behavioral locomotor activity and c-fos expression in the nucleus accumbens and striatum utilizing the immunocytochemical detection of the Fos protein. Nicotine 41-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 197-200 15044039-0 2004 Role of dopamine D1 receptors in the striatal and cortical fos expression induced by the muscarinic agonist pilocarpine. Pilocarpine 108-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 15016420-3 2004 Central sympathectomy with intracerebroventricular injection of 6-OHDA significantly reduced the elevation of the plasma corticosterone level, the expression of Fos in hypothalamic paraventricular nucleus and in locus coeruleus, as well as the suppression of NK activity induced by cold stress at 4 degrees C for 4 h. Peripheral sympathectomy with intraperitoneal (i.p.) Oxidopamine 64-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-164 15044039-1 2004 Injections of the muscarinic cholinergic receptor agonist pilocarpine (50 mg/kg) induced pronounced expression of the immediate early gene (IEG) product Fos in the striatum and cortex of rats. Pilocarpine 58-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-156 15044039-3 2004 In contrast, the muscarinic receptor antagonist scopolamine (0.75-3.00 mg/kg) virtually abolished the Fos response at both sites. Scopolamine 48-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 15033383-5 2004 Using immunohistochemical staining of Fos, present results indicate that the administration of naloxone methiodide or naloxone to morphine-dependent rats induced marked Fos immunoreactivity within the cardiomyocyte nuclei. N-methylnaloxone 95-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 15053967-0 2004 c-fos antisense oligonucleotides increase firing rate of striatal neurons in the anaesthetized rat. Oligonucleotides 16-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 15053967-3 2004 We conclude that, under urethane anesthesia, which here does not affect c-fos expression in the striatum by itself, neuronal activity appears to be tonically suppressed by basal striatal c-fos expression. Urethane 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 187-192 14755517-6 2004 Whereas OVAR-induced Fos-ir neurons were also first observed in vestibular-related brain areas, such as the prepositus hypoglossal nucleus, gigantocellular reticular nucleus, and locus coeruleus, of normal experimental rats at P7, those in the inferior olive were observed only from P14 onward. ovar 8-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 14755517-8 2004 In general, age-dependent increase in OVAR-induced Fos-ir neurons was observed in brain areas that received otolith inputs. ovar 38-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 15033383-0 2004 Morphine withdrawal-induced c-fos expression in the heart: a peripheral mechanism. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 14592947-5 2004 Exposure of the rat gastric mucosa to HCl (0.15-0.5 M) or NH(4)OH (0.1-0.3 M) led to a concentration-dependent expression of c-Fos in the NTS, which was not related to gender, gastric mucosal injury, or gastropyloric motor alterations. Hydrochloric Acid 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 14592947-5 2004 Exposure of the rat gastric mucosa to HCl (0.15-0.5 M) or NH(4)OH (0.1-0.3 M) led to a concentration-dependent expression of c-Fos in the NTS, which was not related to gender, gastric mucosal injury, or gastropyloric motor alterations. Ammonium Hydroxide 58-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 14592947-6 2004 The c-Fos response to HCl was diminished by cimetidine and omeprazole, enhanced by pentagastrin, and left unchanged by dexloxiglumide and itriglumide. Hydrochloric Acid 22-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 14592947-6 2004 The c-Fos response to HCl was diminished by cimetidine and omeprazole, enhanced by pentagastrin, and left unchanged by dexloxiglumide and itriglumide. Cimetidine 44-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 14592947-6 2004 The c-Fos response to HCl was diminished by cimetidine and omeprazole, enhanced by pentagastrin, and left unchanged by dexloxiglumide and itriglumide. Omeprazole 59-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 14592947-6 2004 The c-Fos response to HCl was diminished by cimetidine and omeprazole, enhanced by pentagastrin, and left unchanged by dexloxiglumide and itriglumide. Pentagastrin 83-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 14592947-6 2004 The c-Fos response to HCl was diminished by cimetidine and omeprazole, enhanced by pentagastrin, and left unchanged by dexloxiglumide and itriglumide. dexloxiglumide 119-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 14592947-7 2004 Pentagastrin alone caused an omeprazole-resistant expression of c-fos, which in the NTS was attenuated by itriglumide and prevented by dexloxiglumide but in the area postrema was reduced by dexloxiglumide and abolished by itriglumide. Pentagastrin 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 14592947-7 2004 Pentagastrin alone caused an omeprazole-resistant expression of c-fos, which in the NTS was attenuated by itriglumide and prevented by dexloxiglumide but in the area postrema was reduced by dexloxiglumide and abolished by itriglumide. Omeprazole 29-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 14592947-7 2004 Pentagastrin alone caused an omeprazole-resistant expression of c-fos, which in the NTS was attenuated by itriglumide and prevented by dexloxiglumide but in the area postrema was reduced by dexloxiglumide and abolished by itriglumide. Itriglumide 106-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 14592947-7 2004 Pentagastrin alone caused an omeprazole-resistant expression of c-fos, which in the NTS was attenuated by itriglumide and prevented by dexloxiglumide but in the area postrema was reduced by dexloxiglumide and abolished by itriglumide. dexloxiglumide 135-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 14592947-7 2004 Pentagastrin alone caused an omeprazole-resistant expression of c-fos, which in the NTS was attenuated by itriglumide and prevented by dexloxiglumide but in the area postrema was reduced by dexloxiglumide and abolished by itriglumide. dexloxiglumide 190-204 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 14592947-7 2004 Pentagastrin alone caused an omeprazole-resistant expression of c-fos, which in the NTS was attenuated by itriglumide and prevented by dexloxiglumide but in the area postrema was reduced by dexloxiglumide and abolished by itriglumide. Itriglumide 222-233 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 15123357-10 2004 CCN2-induced c-fos gene activation, expression and cell proliferation were blocked by inhibiting ERK1/2 with PD98059. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 109-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 15049858-7 2004 c-fos mRNA increases following stress were augmented by EB but inhibited by DHTP. dihydrotestosterone propionate 76-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 15103665-10 2004 Fluoxetine not only significantly reduced c-fos expression in all regions in the CMS model in rats, but only showed a marked decrease in c-fos expression in the hippocampus in unstressed animals. Fluoxetine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 15103665-10 2004 Fluoxetine not only significantly reduced c-fos expression in all regions in the CMS model in rats, but only showed a marked decrease in c-fos expression in the hippocampus in unstressed animals. Fluoxetine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 14751419-3 2004 To date, several studies have examined c-fos expression following MDMA administration in rats. N-Methyl-3,4-methylenedioxyamphetamine 66-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 15036599-4 2004 injection of 6-OHDA reduced significantly the elevation of plasma corticosterone level, the expression of Fos in hypothalamic paraventricular nucleus and in locus coeruleus, as well as the suppression of NK activity induced by cold stress at 4 degrees C for 4 h. Peripheral sympathectomy with intraperitoneal (i.p.) Oxidopamine 13-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 14767061-0 2004 c-Fos activated phospholipid synthesis is required for neurite elongation in differentiating PC12 cells. Phospholipids 16-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 14767061-1 2004 We have previously shown that c-Fos activates phospholipid synthesis through a mechanism independent of its genomic AP-1 activity. Phospholipids 46-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 14767061-3 2004 Blocking c-Fos expression inhibited differentiation, phospholipid synthesis activation, and neuritogenesis. Phospholipids 53-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 14767061-5 2004 However, transfected cells expressing c-Fos or c-Fos deletion mutants with capacity to activate phospholipid synthesis sustain neurite outgrowth and elongation in the absence of nerve growth factor. Phospholipids 96-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 14767061-5 2004 However, transfected cells expressing c-Fos or c-Fos deletion mutants with capacity to activate phospholipid synthesis sustain neurite outgrowth and elongation in the absence of nerve growth factor. Phospholipids 96-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 14767061-6 2004 Results disclose a dual function of c-Fos: it first releases the genomic program for differentiation and then associates to the endoplasmic reticulum and activates phospholipid synthesis. Phospholipids 164-176 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 15019966-0 2004 Electroencephalographic, behavioral, and c-fos responses to acute domoic acid exposure. domoic acid 66-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 15019966-8 2004 In postmortem brains obtained immediately after the sessions, c-fos was activated in the anterior olfactory nucleus by both the low and high doses of domoic acid. domoic acid 150-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 15019966-10 2004 These data indicate that electroencephalographic and c-fos responses can be obtained at a dose of domoic acid that fails to activate the behavioral response most commonly used as a bioassay for this marine toxin: ear scratching with the ipsilateral foot. domoic acid 98-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 15094496-3 2004 Formalin induced hind paw inflammation in the rat resulted in bilateral increase of Fos and NADPH-d expression in the pedunculopontine tegmental nucleus. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 15094496-5 2004 Close proximity to NADPH-d labelled neuronal processes with Fos-labelled nuclei were observed. NADP 19-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 15006454-7 2004 The majority of behavioral effects and the elevated c-fos mRNA expression were attenuated by 10 mg/kg DMP696, a CRF(1) antagonist. DMP 696 102-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 15033383-5 2004 Using immunohistochemical staining of Fos, present results indicate that the administration of naloxone methiodide or naloxone to morphine-dependent rats induced marked Fos immunoreactivity within the cardiomyocyte nuclei. N-methylnaloxone 95-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-172 14725963-0 2004 Electro-acupuncture preconditioning abrogates the elevation of c-Fos and c-Jun expression in neonatal hypoxic-ischemic rat brains induced by glibenclamide, an ATP-sensitive potassium channel blocker. Glyburide 141-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 14725963-2 2004 Using Western blot, the expression of c-fos protein (c-Fos) and c-jun protein (c-Jun) induced by glibenclamide, an ATP-sensitive potassium (K(ATP)) channel blocker was examined from cerebral cortical and hippocampal samples in neonatal hypoxic-ischemic rats, with or without EA preconditioning. Glyburide 97-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 15033383-5 2004 Using immunohistochemical staining of Fos, present results indicate that the administration of naloxone methiodide or naloxone to morphine-dependent rats induced marked Fos immunoreactivity within the cardiomyocyte nuclei. Naloxone 95-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 14725963-2 2004 Using Western blot, the expression of c-fos protein (c-Fos) and c-jun protein (c-Jun) induced by glibenclamide, an ATP-sensitive potassium (K(ATP)) channel blocker was examined from cerebral cortical and hippocampal samples in neonatal hypoxic-ischemic rats, with or without EA preconditioning. Glyburide 97-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 14725963-6 2004 Interestingly, low c-Fos and c-Jun expressions were found both in diazoxide and EA groups, 24 h after HI. Diazoxide 66-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 15033383-5 2004 Using immunohistochemical staining of Fos, present results indicate that the administration of naloxone methiodide or naloxone to morphine-dependent rats induced marked Fos immunoreactivity within the cardiomyocyte nuclei. Naloxone 95-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-172 14725963-8 2004 However, the level of c-Fos and c-Jun expression in the group administered glibenclamide after EA was significantly lower than in the glibenclamide group (P< or =0.05). Glyburide 75-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 15033383-5 2004 Using immunohistochemical staining of Fos, present results indicate that the administration of naloxone methiodide or naloxone to morphine-dependent rats induced marked Fos immunoreactivity within the cardiomyocyte nuclei. Morphine 130-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 14725964-0 2004 Morphine- and cocaine-induced c-Fos levels in Lewis and Fischer rat strains. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 15033383-5 2004 Using immunohistochemical staining of Fos, present results indicate that the administration of naloxone methiodide or naloxone to morphine-dependent rats induced marked Fos immunoreactivity within the cardiomyocyte nuclei. Morphine 130-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-172 14725964-0 2004 Morphine- and cocaine-induced c-Fos levels in Lewis and Fischer rat strains. Cocaine 14-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 14725964-8 2004 The present results indicated that patterns of morphine- and cocaine-induced c-Fos within CTA-associated, but not reward- or locomotor-associated, brain regions paralleled the differential behavioral sensitivities of LEW and F344 rats to these drugs within CTA learning. Morphine 47-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 15033383-6 2004 Moreover, Western blot analysis revealed a peak expression of c-fos in the right and left ventricles after naloxone methiodide- or naloxone-precipitated withdrawal. N-methylnaloxone 107-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 14725964-8 2004 The present results indicated that patterns of morphine- and cocaine-induced c-Fos within CTA-associated, but not reward- or locomotor-associated, brain regions paralleled the differential behavioral sensitivities of LEW and F344 rats to these drugs within CTA learning. Cocaine 61-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 15033383-6 2004 Moreover, Western blot analysis revealed a peak expression of c-fos in the right and left ventricles after naloxone methiodide- or naloxone-precipitated withdrawal. Naloxone 107-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 15033383-7 2004 In addition, in the hypothalamic paraventricular nucleus (PVN), Fos expression was increased after naloxone-but not after naloxone methiodide-administration to morphine-dependent rats. Naloxone 99-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 15033383-8 2004 These results suggest that the activation of c-fos expression observed during morphine withdrawal in the heart is due to intrinsic mechanisms outside the central nervous system (CNS). Morphine 78-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 15106833-2 2004 The present study examined Fos expression associated with pain-related aversion in rats, using formalin-induced conditioned place avoidance (F-CPA) test, which could distinguish pain emotion from pain sensation. Formaldehyde 95-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 15084346-5 2004 Maximum levels of the c-fos and VEGF mRNA after raloxifene treatment were higher than those seen after treatments with E2 or a corresponding dose of tamoxifen or ospemifene. Raloxifene Hydrochloride 48-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 14742673-0 2004 Nitric oxide regulates c-fos expression in nucleus tractus solitarii induced by baroreceptor activation via cGMP-dependent protein kinase and cAMP response element-binding protein phosphorylation. Nitric Oxide 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 14742673-0 2004 Nitric oxide regulates c-fos expression in nucleus tractus solitarii induced by baroreceptor activation via cGMP-dependent protein kinase and cAMP response element-binding protein phosphorylation. Cyclic AMP 142-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 14742673-6 2004 Bilateral NTS microinjection of a cell-permeable cGMP analog, 8-bromoguanosine-3",5"-cyclic monophosphate (10 nmol) significantly elevated the level of pCREB or c-fos mRNA in the NTS. Cyclic GMP 49-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 14742673-6 2004 Bilateral NTS microinjection of a cell-permeable cGMP analog, 8-bromoguanosine-3",5"-cyclic monophosphate (10 nmol) significantly elevated the level of pCREB or c-fos mRNA in the NTS. 8-bromocyclic GMP 62-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 14742673-8 2004 We conclude that NO derived from nNOS in the NTS on baroreceptor activation may participate in c-fos expression via phosphorylation of CREB in a process that engages the sGC/cGMP/PKG-I signaling cascade. Cyclic GMP 174-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 14745483-0 2004 Fos immunocytochemical studies on the neuroanatomical sites of action of acute tyrosine depletion in the rat brain. Tyrosine 79-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 14745483-3 2004 OBJECTIVES: Here we used fos immunocytochemistry to examine the neuroanatomical sites of action of a tyrosine-free amino acid mixture administered either alone or combined with amphetamine. Tyrosine 101-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 14745483-9 2004 However, the mixture reduced the increase in fos expression evoked by amphetamine. Amphetamine 70-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 14745483-12 2004 CONCLUSIONS: In summary, a tyrosine-free amino acid mixture reduced amphetamine-induced fos expression but this effect was region-specific and included dopamine-rich regions. Tyrosine 27-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 14745483-12 2004 CONCLUSIONS: In summary, a tyrosine-free amino acid mixture reduced amphetamine-induced fos expression but this effect was region-specific and included dopamine-rich regions. Amphetamine 68-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 14749430-4 2004 During perforant pathway stimulation in urethane-anesthetized rats, passive diffusion of the GABA(A) receptor antagonist bicuculline methiodide from the tip of a glass recording electrode evoked granule cell discharges and c-Fos expression in granule cells, mossy cells, and inhibitory interneurons, within a approximately 400 microm radius. bicuculline methiodide 121-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 223-228 14689487-9 2004 Anesthesia with halothane induced the strongest c-Fos expression in a restricted pool of pmXII located in the pons at the level of the Kolliker-Fuse nucleus and the intertrigeminal region. Halothane 16-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 14709343-1 2004 Acute administration of morphine induces expression of the immediate-early gene (IEG) c-Fos in dorsomedial striatum, portions of cerebral cortex, and in several midline-intralaminar thalamic nuclei, partly via a trans-synaptic mechanism that involves activation of glutamate receptors. Morphine 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 14709343-2 2004 Because activation of protein kinase C (PKC) may occur following the activation of glutamate receptors, we determined whether pharmacological inhibition of PKC would attenuate morphine-induced c-Fos expression, and whether acute administration of morphine would induce translocation of PKC. Morphine 176-184 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-198 14709343-3 2004 The selective PKC antagonist NPC 15437 given 30 min prior to morphine significantly decreased morphine-induced c-Fos expression in striatum and cingulate cortex, but not in centrolateral thalamus. Morphine 94-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 14709343-8 2004 Double immunocytochemistry at the light microscopic level demonstrated co-localization of translocated PKC betaII and c-Fos in some cortical neurons 90 min after morphine injection. Morphine 162-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 12958059-8 2004 Pretreatment with the amylin antagonist AC-187 (1 mg/kg sc) inhibited feeding-induced c-Fos expression in the AP. AC 187 40-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 14565991-9 2004 However, in the presence of CsA, c-fos mRNA levels were significantly augmented by increased pacing frequency. Cyclosporine 28-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 14592853-6 2004 Interestingly, co-administration of nicotine and Ang II at lower doses, which did not affect cell growth, induced DNA synthesis and c-fos expression accompanied by enhancement of ERK, STAT, and p38MAPK activity. Nicotine 36-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 15630176-1 2004 Two key properties of short chain fructooligosaccharides (sc-FOS) which lead to physiological functions are indigestibility in the small intestine and fermentability in the colon. fructooligosaccharide 34-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 14706869-2 2004 In the present study, serial unilateral transtympanic administration of TTX produced behavioral symptoms indicative of transient vestibular disruption and novel patterns of Fos activity in the brainstem and cerebellum. Tetrodotoxin 72-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 173-176 14706869-3 2004 Following two or three serial injections of TTX and a final survival time of 2 h, Fos immunocytochemistry revealed a distinct pattern of labeling in the brainstem that differed temporally from that observed following a single unilateral TTX injection. Tetrodotoxin 44-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 14706869-4 2004 Specifically there was protracted expression of Fos in the beta subdivision of the inferior olive (IO) on the side ipsilateral to TTX treatment. Tetrodotoxin 130-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 14706869-6 2004 In particular, there was prominent Fos labeling of Purkinje cells in the contra-TTX half of lobule X. Tetrodotoxin 80-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 15630176-2 2004 Sc-FOS is converted into short chain fatty acids (SCFAs) by intestinal bacteria in the colon and absorbed. Fatty Acids, Volatile 25-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 3-6 15630176-4 2004 Besides improvement of GI condition, dietary sc-FOS influences on calcium and magnesium absorption in the colon. Calcium 66-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 15630176-4 2004 Besides improvement of GI condition, dietary sc-FOS influences on calcium and magnesium absorption in the colon. Magnesium 78-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 15630176-5 2004 A major mineral absorption site is the small intestine, but the colon also works as a Ca and Mg absorption site with an aid of SCFAs made from sc-FOS. Magnesium 93-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-149 15630176-6 2004 Furthermore dietary sc-FOS influences on bioavailability of soy-isoflavones. Isoflavones 60-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 14725632-1 2004 Gamma-amino butyric acid (GABA)A receptor stimulation in the nucleus accumbens shell produces intense hyperphagia in rats and increases Fos expression in the lateral hypothalamus. gamma-Aminobutyric Acid 0-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-139 14725632-5 2004 Intra-accumbens shell muscimol treatment significantly increased the percentage of orexin/hypocretin-containing neurons expressing Fos in the lateral, but not medial, portion of the perifornical/lateral hypothalamic area. Muscimol 22-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-134 14725632-8 2004 Muscimol treatment also markedly increased Fos expression in the arcuate nucleus, which connects reciprocally to the lateral/perifornical hypothalamic area. Muscimol 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 14750966-3 2004 Two hours after intragastric treatment with HCl or NH4OH the activation of neurons in the nucleus tractus solitarii at the rostrocaudal extension of the area postrema (NTSAP) was visualized by c-Fos immunohistochemistry and their chemical coding characterized by double-labelling immunohistochemistry. Hydrochloric Acid 44-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-198 14750966-3 2004 Two hours after intragastric treatment with HCl or NH4OH the activation of neurons in the nucleus tractus solitarii at the rostrocaudal extension of the area postrema (NTSAP) was visualized by c-Fos immunohistochemistry and their chemical coding characterized by double-labelling immunohistochemistry. Ammonium Hydroxide 51-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-198 14750966-4 2004 Exposure of the rat gastric mucosa to HCl (0.15-0.5 M) or NH4OH (0.1-0.3 M) led to a concentration-dependent expression of c-Fos in the NTSAP. Hydrochloric Acid 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 14750966-4 2004 Exposure of the rat gastric mucosa to HCl (0.15-0.5 M) or NH4OH (0.1-0.3 M) led to a concentration-dependent expression of c-Fos in the NTSAP. Ammonium Hydroxide 58-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 14750966-7 2004 The similar number and distribution of c-Fos-expressing neurons within the NTSAP and their identical chemical coding indicate that exposure of the rat stomach to backdiffusing concentrations of HCl and NH4OH activates the same vagal afferent-NTSAP pathway. Hydrochloric Acid 194-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 14750966-7 2004 The similar number and distribution of c-Fos-expressing neurons within the NTSAP and their identical chemical coding indicate that exposure of the rat stomach to backdiffusing concentrations of HCl and NH4OH activates the same vagal afferent-NTSAP pathway. Ammonium Hydroxide 202-207 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 14642828-10 2003 In areas outside the NTS, such as the dorsal motor nucleus of the vagus (DMV), Fos activation was absent despite a dense oxytocin and CRH innervation. (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 73-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 15630176-9 2004 These results suggest that FOS increase the bioavailability of isoflavones. Isoflavones 63-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 15382258-0 2004 Fos induction and persistence, neurodegeneration, and interneuron activation in the hippocampus of epilepsy-resistant versus epilepsy-prone rats after pilocarpine-induced seizures. Pilocarpine 151-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 14997016-3 2004 To study the hypothesis that suppression of Fos in the shell is a correlate of photic entrainment, we used rats that were treated with the retinal neurotoxin monosodium glutamate (MSG) during the neonatal period. Sodium Glutamate 158-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 15173632-0 2004 Harmaline-induced climbing fiber activation causes amino acid and peptide release in the rodent cerebellar cortex and a unique temporal pattern of Fos expression in the olivo-cerebellar pathway. Harmaline 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-150 15173632-5 2004 Fos expression was first detected in the inferior olive at 15 min post-harmaline injection followed by expression in the deep cerebellar nuclei (30 min) and then in the cerebellar cortex (1 h). Harmaline 71-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 15050102-4 2004 For the purpose of the study, c-Fos-IR has been analysed by immunohistochemical methods in rat pancreatic beta cells, pulpal undifferentiated ectomesenchimal cells (PUECs) that are known to have a neural crest origin, and in small intestine fibroblasts which do not have a neural crest origin, in formaline-fixed, paraffin-embedded sections. Formaldehyde 297-306 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 15050102-4 2004 For the purpose of the study, c-Fos-IR has been analysed by immunohistochemical methods in rat pancreatic beta cells, pulpal undifferentiated ectomesenchimal cells (PUECs) that are known to have a neural crest origin, and in small intestine fibroblasts which do not have a neural crest origin, in formaline-fixed, paraffin-embedded sections. Paraffin 314-322 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 14583742-9 2004 CCK-8s injected into the dPAG induced Fos immunoreactivity in several brain areas related to defensive behavior, including the PAG, median, and dorsal raphe nuclei, superior colliculus, lateral septal nuclei, medial hypothalamus, and medial amygdala. dpag 25-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 14583742-9 2004 CCK-8s injected into the dPAG induced Fos immunoreactivity in several brain areas related to defensive behavior, including the PAG, median, and dorsal raphe nuclei, superior colliculus, lateral septal nuclei, medial hypothalamus, and medial amygdala. phenylacetylglycine 26-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 15489025-3 2004 Additionally, we investigated if moxonidine into the LPBN would modify furosemide+captopril-induced c-fos expression in the forebrain. moxonidine 33-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 15489025-3 2004 Additionally, we investigated if moxonidine into the LPBN would modify furosemide+captopril-induced c-fos expression in the forebrain. Furosemide 71-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 15489025-3 2004 Additionally, we investigated if moxonidine into the LPBN would modify furosemide+captopril-induced c-fos expression in the forebrain. Captopril 82-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 15501601-7 2004 administration of interleukin-1beta and tumor necrosis factor-alpha alone induced c-Fos in the brainstem, an effect that was gone after 24 h. At this time, however, the effect of HCl to cause expression of c-Fos in the nucleus tractus solitarii was significantly enhanced by pretreatment with interleukin-1beta and tumor necrosis factor-alpha. Hydrochloric Acid 179-182 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 15501601-7 2004 administration of interleukin-1beta and tumor necrosis factor-alpha alone induced c-Fos in the brainstem, an effect that was gone after 24 h. At this time, however, the effect of HCl to cause expression of c-Fos in the nucleus tractus solitarii was significantly enhanced by pretreatment with interleukin-1beta and tumor necrosis factor-alpha. Hydrochloric Acid 179-182 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 206-211 13680075-14 2004 (iii) The agreement between mirtazapine and imipramine increases our confidence in the validity of c-fos expression profiling to aid drug classification and predict therapeutic utility. Mirtazapine 28-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 13680075-14 2004 (iii) The agreement between mirtazapine and imipramine increases our confidence in the validity of c-fos expression profiling to aid drug classification and predict therapeutic utility. Imipramine 44-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 14695781-5 2004 HGJs could be blocked with carbenoxolone (CBX), a gap junction blocker, and the number of Fos-LI neurons was significantly decreased compared with that in rats without CBX injection, while Fos-LI ASs were not affected. Carbenoxolone 42-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 189-192 14741756-0 2003 c-fos and cleaved caspase-3 expression after perinatal exposure to ethanol, cocaine, or the combination of both drugs. Ethanol 67-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 14741756-0 2003 c-fos and cleaved caspase-3 expression after perinatal exposure to ethanol, cocaine, or the combination of both drugs. Cocaine 76-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 14741756-5 2003 At birth, the brains of fetuses exposed to cocaine exhibited an increase in Fos immunoreactivity in many brain regions. Cocaine 43-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 14741756-7 2003 However, Fos expression at 24 h after birth was higher after ethanol diet treatment in several brain regions, such as the amygdala, ventromedial hypothalamus, and medial thalamus. Ethanol 61-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 14741756-8 2003 Only in the striatum did the combination of ethanol and cocaine cause greater Fos expression than either prenatal cocaine or ethanol alone. Ethanol 44-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 14741756-8 2003 Only in the striatum did the combination of ethanol and cocaine cause greater Fos expression than either prenatal cocaine or ethanol alone. Cocaine 56-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 14741756-12 2003 These results indicate that both prenatal cocaine and prenatal ethanol exposure increase Fos and cleaved caspase-3 expression in the developing brain in a time- and region-dependent manner, but that the combination of low-dose, chronic ethanol, and binge cocaine does not cause greater apoptosis. Cocaine 42-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 14741756-12 2003 These results indicate that both prenatal cocaine and prenatal ethanol exposure increase Fos and cleaved caspase-3 expression in the developing brain in a time- and region-dependent manner, but that the combination of low-dose, chronic ethanol, and binge cocaine does not cause greater apoptosis. Ethanol 63-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 12933360-4 2004 Diurnal patterns of c-Fos-IR were observed in the tuberomammilar nucleus (TM) and suprachiasmatic nucleus (SCN) in AL rats. Aluminum 115-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 12933360-7 2004 Food-entrained c-Fos-IR patterns persisted after 3 days in fasting in DMH, LH, and PeF. Dimenhydrinate 70-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 15541892-0 2004 Cocaine pre-exposure produces a sensitized and context-specific c-fos mRNA response to footshock stress in the central nucleus of the AMYGDALA. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 15541892-10 2004 Furthermore, this effect was gated by the environmental context in which cocaine was given; footshock only enhanced c-fos mRNA expression when it was given in a context that had previously been paired with cocaine. Cocaine 73-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 15541892-10 2004 Furthermore, this effect was gated by the environmental context in which cocaine was given; footshock only enhanced c-fos mRNA expression when it was given in a context that had previously been paired with cocaine. Cocaine 206-213 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 14667454-4 2004 The decrease of light-induced Fos expression in OLETF rats was significantly reversed by pretreatment with the 5-HT1B receptor antagonist, isamoltan (3 mg/kg, i.p.). isamoltane 139-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 14667454-6 2004 ), a 5-HT1B agonist, blocked the reversal effect of isamoltan on Fos expression. isamoltane 52-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 14751279-0 2004 Long-term behavioral and neuronal cross-sensitization to amphetamine induced by repeated brief social defeat stress: Fos in the ventral tegmental area and amygdala. Amphetamine 57-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 14751279-9 2004 Amphetamine augmented stress-induced Fos-LI labeling 17 days after the first stress episode in the dorsal striatum, NAc core, and medial amygdala, reflecting a cross-sensitization of Fos response. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 14751279-9 2004 Amphetamine augmented stress-induced Fos-LI labeling 17 days after the first stress episode in the dorsal striatum, NAc core, and medial amygdala, reflecting a cross-sensitization of Fos response. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 183-186 14751279-10 2004 Amphetamine challenge 70 days after social stress exposures revealed sensitized Fos-LI labeling in the VTA and the amygdala. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 14960337-8 2004 Examination of neuronal activation, as indexed by the immediate early gene c-fos, showed that A 68930 and dihydrexidine caused a highly significant expression of c-fos in the medial prefrontal cortex. dihydrexidine 106-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 14960337-8 2004 Examination of neuronal activation, as indexed by the immediate early gene c-fos, showed that A 68930 and dihydrexidine caused a highly significant expression of c-fos in the medial prefrontal cortex. dihydrexidine 106-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 14960337-10 2004 The effects of A 68930 and dihydrexidine on c-fos expression in caudate putamen or nucleus accumbens were less marked, or undetectable. dihydrexidine 27-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 14980739-8 2004 stimulated Fos expression in the ARC, ADP, and LA. Adenosine Diphosphate 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 15099693-0 2004 Response to kainic acid injections: changes in staining for zinc, FOS, cell death and glial response in the rat forebrain. Kainic Acid 12-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 15099693-10 2004 Transitory Fos immunostaining (within neuronal nuclei) was observed between 3 and 12 h after kainate treatment in many telencephalic areas: olfactory bulb, cortex (piriform, hippocampal and neocortex) and amygdaloid nuclei. Kainic Acid 93-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 15183509-0 2004 Repeated application of ketamine to rats induces changes in the hippocampal expression of parvalbumin, neuronal nitric oxide synthase and cFOS similar to those found in human schizophrenia. Ketamine 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-142 15183518-5 2004 In the NAc shell, morphine administration resulted in upregulation of caspace 9, NF-kappaB, NF-H, tau, GABA-A delta subunit, FGFR1, Ggamma2, synuclein 1, syntaxin 5 and 13, GRK5, and c-fos mRNAs. Morphine 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 183-188 15219685-0 2004 Cocaine-induced Fos expression in rat striatum is blocked by chloral hydrate or urethane. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 15219685-0 2004 Cocaine-induced Fos expression in rat striatum is blocked by chloral hydrate or urethane. Chloral Hydrate 61-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 15219685-0 2004 Cocaine-induced Fos expression in rat striatum is blocked by chloral hydrate or urethane. Urethane 80-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 15219685-4 2004 We first assayed the ability of 20 mg/kg cocaine to induce Fos expression in the striatum following pretreatment with 400 mg/kg chloral hydrate or 1.3 g/kg urethane, two of the most commonly used anesthetics for in vivo electrophysiology. Cocaine 41-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 15219685-5 2004 Chloral hydrate blocked, while urethane strongly attenuated cocaine-induced Fos expression without affecting basal levels of expression. Urethane 31-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 15219685-5 2004 Chloral hydrate blocked, while urethane strongly attenuated cocaine-induced Fos expression without affecting basal levels of expression. Cocaine 60-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 15219685-6 2004 We then examined dopaminergic and glutamatergic mechanisms for anesthetic effects on cocaine-induced Fos expression. Cocaine 85-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-104 15219685-11 2004 To assess a causal role for a reduction of glutamatergic neurotransmission in anesthetic effects on cocaine-induced Fos expression, we injected the glutamate receptor agonists AMPA and NMDA into the dorsal striatum of chloral hydrate-anesthetized rats. Cocaine 100-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 15219685-12 2004 The glutamate receptor agonists partially reinstated cocaine-induced Fos expression in anesthetized rats. Cocaine 53-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 14674993-0 2004 Expressions of junB and c-fos are enhanced in 4-nitroquinoline 1-oxide-induced rat tongue cancers. 4-Nitroquinoline 46-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 14674993-0 2004 Expressions of junB and c-fos are enhanced in 4-nitroquinoline 1-oxide-induced rat tongue cancers. 1-oxide 63-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 14695781-4 2004 The number of HGJs and Fos like immunoreactive (-LI) cells was significantly increased following hyperosmotic stimuli, that is, the rats were administered 30 g/L NaCl solution orally or 90 g/L NaCl solution intravenously. nacl solution 162-175 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 14695781-4 2004 The number of HGJs and Fos like immunoreactive (-LI) cells was significantly increased following hyperosmotic stimuli, that is, the rats were administered 30 g/L NaCl solution orally or 90 g/L NaCl solution intravenously. nacl solution 193-206 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 14695781-5 2004 HGJs could be blocked with carbenoxolone (CBX), a gap junction blocker, and the number of Fos-LI neurons was significantly decreased compared with that in rats without CBX injection, while Fos-LI ASs were not affected. Carbenoxolone 27-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 14695781-5 2004 HGJs could be blocked with carbenoxolone (CBX), a gap junction blocker, and the number of Fos-LI neurons was significantly decreased compared with that in rats without CBX injection, while Fos-LI ASs were not affected. Carbenoxolone 27-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 189-192 14695781-5 2004 HGJs could be blocked with carbenoxolone (CBX), a gap junction blocker, and the number of Fos-LI neurons was significantly decreased compared with that in rats without CBX injection, while Fos-LI ASs were not affected. Carbenoxolone 42-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 14663201-0 2003 Regional pattern of binding and c-Fos induction by (R)- and (S)-citalopram in rat brain. (r)- and (s)-citalopram 51-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 14663201-5 2003 S-citalopram induced a significant increase in c-Fos positive cells in central amygdala and in the bed nucleus of the stria terminalis (BST), compared to vehicle controls. Citalopram 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 14566971-5 2003 PD98059, a MEK inhibitor, attenuated AP-1 activation, lowered c-Fos and Fra-1 protein levels and reduced cell number and cells positive for proliferating cell nuclear antigen in old. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 15075448-6 2003 The gastric acid-induced expression of c-fos in the brainstem is reduced by inhibition of gastric acid secretion and enhanced by pentagastrin-evoked stimulation of gastric acid secretion. Pentagastrin 129-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 12955385-0 2003 Interaction between the noradrenergic and serotonergic systems in locomotor hyperactivity and striatal expression of Fos induced by amphetamine in rats. Amphetamine 132-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 14663201-6 2003 A similar pattern of c-Fos activation was observed with (RS)-citalopram, sertraline and paroxetine, while (R)-citalopram did not increase c-Fos expression in these areas. Sertraline 73-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 14663201-6 2003 A similar pattern of c-Fos activation was observed with (RS)-citalopram, sertraline and paroxetine, while (R)-citalopram did not increase c-Fos expression in these areas. Paroxetine 88-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 14563495-0 2003 Fangchinoline inhibits rat aortic vascular smooth muscle cell proliferation and cell cycle progression through inhibition of ERK1/2 activation and c-fos expression. fangchinoline 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-152 14517181-5 2003 To investigate the importance of glucose availability, we studied the effect of CCK receptor antagonists on c-Fos synthesis induced by the glucose antimetabolite 2-deoxyglucose. Deoxyglucose 162-176 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 14515343-6 2003 In parallel studies, the same dose of fenfluramine was shown to be sufficient to cause an increase in the expression of the immediate early genes (IEG) c-fos and Arc mRNA in cortical regions with high 5-HT(2A) receptor density. Fenfluramine 38-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-157 14600289-0 2003 Hexachlorobenzene-induced early changes in ornithine decarboxylase and protein tyrosine kinase activities, polyamines and c-Myc, c-Fos and c-Jun proto-oncogenes in rat liver. Hexachlorobenzene 0-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 14600289-4 2003 HCB (1000 mg/kg body weight) increased hepatic immunodetectable c-Myc, c-Fos, and c-Jun levels after 6 h, and ODC activity and spermine and putrescine content after 18 and 24 h, while maximum stimulation of PTK activity occurred at 12 h. PTK and ODC activities varied in a dose-dependent manner. Hexachlorobenzene 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 14600289-6 2003 They were all elevated at the second day of treatment, while only c-Fos and c-Jun remained elevated after 10 days of HCB exposure. Hexachlorobenzene 117-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 14645470-0 2003 Opioid modulation of scratching and spinal c-fos expression evoked by intradermal serotonin. Serotonin 82-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 14645470-4 2003 Intradermal 5-HT elicited a significant increase in c-fos-like immunoreactivity (FLI) in superficial laminas I-III at the lateral aspect of the cervical C3-C6 dorsal horn compared with controls receiving intradermal saline. Sodium Chloride 216-222 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 14627632-11 2003 Furthermore, estradiol selectively affected the expression of c-Fos mRNA and protein by attenuating the injury-induced increase in a time- and region-specific manner. Estradiol 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 14596890-4 2003 In the immunocytochemical study, the post-conditioning administration of AIDA decreased the c-Fos induction in the dentate gyrus and CA1 layer of the hippocampus proper, 2h after exposure of animals to the aversive context, and 24h after conditioning session. Deuterium 170-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 14596893-2 2003 To this purpose, the effect of the administration of MK-801, an NMDA receptor antagonist, on Zif/268 and c-Fos expression following a noxious stimulus, represented by formalin injection into the whisker pad of rats, was examined in neurons of the trigeminal nucleus caudalis. Dizocilpine Maleate 53-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 14596893-4 2003 Zif/268 or c-Fos immunoreactivity elicited by formalin injection was significantly reduced by pretreatment with MK-801 in the superficial layer of the trigeminal nucleus caudalis; more than 40% of the neurons expressing Zif/268 and c-Fos in this layer were also immunolabeled by NR1. Formaldehyde 46-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 14596893-4 2003 Zif/268 or c-Fos immunoreactivity elicited by formalin injection was significantly reduced by pretreatment with MK-801 in the superficial layer of the trigeminal nucleus caudalis; more than 40% of the neurons expressing Zif/268 and c-Fos in this layer were also immunolabeled by NR1. Formaldehyde 46-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 232-237 14596893-4 2003 Zif/268 or c-Fos immunoreactivity elicited by formalin injection was significantly reduced by pretreatment with MK-801 in the superficial layer of the trigeminal nucleus caudalis; more than 40% of the neurons expressing Zif/268 and c-Fos in this layer were also immunolabeled by NR1. Dizocilpine Maleate 112-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 14596893-4 2003 Zif/268 or c-Fos immunoreactivity elicited by formalin injection was significantly reduced by pretreatment with MK-801 in the superficial layer of the trigeminal nucleus caudalis; more than 40% of the neurons expressing Zif/268 and c-Fos in this layer were also immunolabeled by NR1. Dizocilpine Maleate 112-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 232-237 12955385-5 2003 In normal rats, Amphetamine induced locomotor hyperactivity and striatal expression of Fos. Amphetamine 16-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 12955385-6 2003 Pretreatment with the alpha1-adrenergic-receptor antagonist Prazosin or lesion of the serotonergic system significantly reduced the locomotor hyperactivity and striatal Fos expression induced by Amphetamine. Prazosin 60-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-172 12955385-6 2003 Pretreatment with the alpha1-adrenergic-receptor antagonist Prazosin or lesion of the serotonergic system significantly reduced the locomotor hyperactivity and striatal Fos expression induced by Amphetamine. Amphetamine 195-206 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-172 12955387-6 2003 CGX-1007 treatment reduced c-fos expression throughout the infarct region by up to 50%. N4-cyclopropyl-5-ethyl-6-piperidin-1-yl-pyrimidine-2,4-diamine 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 14597323-8 2003 Regional glucose uptake metabolic mapping and FOS expression studies suggested that striatal dopamine denervation produced increased sensitivity of Group III mGluRs. Dopamine 93-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 14517181-5 2003 To investigate the importance of glucose availability, we studied the effect of CCK receptor antagonists on c-Fos synthesis induced by the glucose antimetabolite 2-deoxyglucose. Glucose 139-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 14622438-4 2003 In the following studies, we observed the effect of acute glucocorticoid (RU28362) treatment on subsequent HPA axis reactivity by measuring stress-induced plasma hormone concentration [corticosterone and adrenocorticotropic hormone (ACTH)] and gene expression (c-fos and CRH) in the PVN. 11,17-dihydroxy-6-methyl-17-(1-propynyl)androsta-1,4,6-triene-3-one 74-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 261-266 14574439-0 2003 Different pattern of brain c-Fos expression following re-exposure to ethanol or sucrose self-administration environment. Ethanol 69-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 14574439-0 2003 Different pattern of brain c-Fos expression following re-exposure to ethanol or sucrose self-administration environment. Sucrose 80-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 14574439-4 2003 c-Fos protein expression was used as a marker of neuronal activation.Re-exposure to ethanol self-administration environment after 30-day but not after 24-h abstinence increased the number of Fos-positive nuclei in the thalamic paraventricular nucleus, granular insular cortex and medial prefrontal cortex. Ethanol 84-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 14574439-4 2003 c-Fos protein expression was used as a marker of neuronal activation.Re-exposure to ethanol self-administration environment after 30-day but not after 24-h abstinence increased the number of Fos-positive nuclei in the thalamic paraventricular nucleus, granular insular cortex and medial prefrontal cortex. Ethanol 84-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 2-5 14550853-0 2003 Naloxone potentiates treadmill running-induced increase in c-Fos expression in rat hippocampus. Naloxone 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 14550853-3 2003 It was shown that c-Fos expression in the CA1 region, the CA2 and CA3 regions, and the dentate gyrus of the hippocampus was significantly increased by treadmill running and naloxone, a nonselective opioid receptors antagonist, treatment enhanced treadmill exercise-induced increase of hippocampal c-Fos expression. Naloxone 173-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 14550853-3 2003 It was shown that c-Fos expression in the CA1 region, the CA2 and CA3 regions, and the dentate gyrus of the hippocampus was significantly increased by treadmill running and naloxone, a nonselective opioid receptors antagonist, treatment enhanced treadmill exercise-induced increase of hippocampal c-Fos expression. Naloxone 173-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 297-302 14614962-4 2003 The number of c-Fos-ir neurons in the nucleus tractus solitarius in response to phenylephrine-induced baroreceptor activation was used as an index of the integrity of the afferent limb of the baroreceptor reflex. Phenylephrine 80-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 14614962-7 2003 alpha-Lipoic acid-treated diabetic rats had plasma malondialdehyde levels similar to those seen in non-diabetic rats and less than those of vehicle-treated diabetic rats at both the 8- and 16-week time points.alpha-Lipoic acid treatment did not affect the baseline (absence of baroreceptor activation) presence of c-Fos-ir in the nucleus tractus solitarius. Thioctic Acid 0-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 314-319 14614962-9 2003 As previously reported, phenylephrine-induced baroreceptor activation resulted in significantly fewer c-Fos-ir neurons in the nucleus tractus solitarius (commissural and caudal subpostremal regions) of diabetic rats when compared to non-diabetic rats at both 8- and 16-week time points. Phenylephrine 24-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 14614962-10 2003 Four weeks of alpha-lipoic acid treatment reversed the diabetes-induced decrement in the numbers of c-Fos-ir neurons in the nucleus tractus solitarius in response to baroreceptor activation. Thioctic Acid 14-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 14614962-11 2003 alpha-Lipoic acid-treated diabetic rats showed the same phenylephrine-induced c-Fos response in the nucleus tractus solitarius as those of alpha-lipoic-acid- and vehicle-treated control rats at both 8- and 16-week time points. Thioctic Acid 0-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 14614962-11 2003 alpha-Lipoic acid-treated diabetic rats showed the same phenylephrine-induced c-Fos response in the nucleus tractus solitarius as those of alpha-lipoic-acid- and vehicle-treated control rats at both 8- and 16-week time points. Phenylephrine 56-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 14584105-1 2003 LiCl at doses sufficient to induce conditioned taste aversion (CTA) causes c-Fos expression in the brain regions implicated in CTA formation. Lithium Chloride 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 14584105-1 2003 LiCl at doses sufficient to induce conditioned taste aversion (CTA) causes c-Fos expression in the brain regions implicated in CTA formation. Chlorotrianisene 63-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 14584105-3 2003 In this study, we examined the effect of central Nomega-nitro-L- arginine methyl ester (L-NAME) on the brain c-Fos expression and CTA learning induced by lithium in rats. NG-Nitroarginine Methyl Ester 49-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 14584105-3 2003 In this study, we examined the effect of central Nomega-nitro-L- arginine methyl ester (L-NAME) on the brain c-Fos expression and CTA learning induced by lithium in rats. NG-Nitroarginine Methyl Ester 88-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 14584105-3 2003 In this study, we examined the effect of central Nomega-nitro-L- arginine methyl ester (L-NAME) on the brain c-Fos expression and CTA learning induced by lithium in rats. Lithium 154-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 14566407-5 2003 (2) Formalin-induced sigmoid pain scores and the expression of Fos in the spinal cord at S1 segment were decreased after microinjecting L-NAME into the DR. Formaldehyde 4-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 14566407-5 2003 (2) Formalin-induced sigmoid pain scores and the expression of Fos in the spinal cord at S1 segment were decreased after microinjecting L-NAME into the DR. NG-Nitroarginine Methyl Ester 136-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 14499961-11 2003 The cannabinoid agonist suppressed formalin-evoked Fos protein expression, a marker of neuronal activity, in the lumbar dorsal horn of sham-operated rats, but no suppression was observed in lesioned rats. Cannabinoids 4-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 14499961-11 2003 The cannabinoid agonist suppressed formalin-evoked Fos protein expression, a marker of neuronal activity, in the lumbar dorsal horn of sham-operated rats, but no suppression was observed in lesioned rats. Formaldehyde 35-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 14499961-12 2003 The number of formalin-evoked Fos-like immunoreactive (FLI) cells was greater in lamina I and II of lesioned rats relative to sham-operated rats. Formaldehyde 14-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 14499961-13 2003 These data indicate that the suppressive effect of the cannabinoid on formalin-evoked Fos protein expression in the superficial dorsal horn was attenuated following destruction of descending noradrenergic pathways. Cannabinoids 55-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 14499961-13 2003 These data indicate that the suppressive effect of the cannabinoid on formalin-evoked Fos protein expression in the superficial dorsal horn was attenuated following destruction of descending noradrenergic pathways. Formaldehyde 70-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 14529807-0 2003 Selective induction of c-Fos immunoreactivity in the prelimbic cortex during reinstatement of heroin seeking induced by acute food deprivation in rats. Heroin 94-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 14529807-11 2003 Food deprivation selectively increased c-Fos immunoreactivity (IR) in the prelimbic cortex of heroin-trained, but not saline-trained, rats (n=4 per condition). Heroin 94-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 14529807-12 2003 Food deprivation also increased c-Fos IR in both heroin- and saline-trained rats in the basolateral amygdala and the ventrolateral bed nucleus of stria terminalis (BNST), but had no effect on c-Fos expression in the dorsolateral BNST, cingulate cortex, nucleus accumbens, and central amygdala. Heroin 49-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 14529807-12 2003 Food deprivation also increased c-Fos IR in both heroin- and saline-trained rats in the basolateral amygdala and the ventrolateral bed nucleus of stria terminalis (BNST), but had no effect on c-Fos expression in the dorsolateral BNST, cingulate cortex, nucleus accumbens, and central amygdala. Sodium Chloride 61-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 14529807-12 2003 Food deprivation also increased c-Fos IR in both heroin- and saline-trained rats in the basolateral amygdala and the ventrolateral bed nucleus of stria terminalis (BNST), but had no effect on c-Fos expression in the dorsolateral BNST, cingulate cortex, nucleus accumbens, and central amygdala. Sodium Chloride 61-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 192-197 12949783-5 2003 In the ventral portion of the trigeminal subnuclei interpolaris/caudalis (Vi/Vc) transition zone, the percentage of Fos-positive neurons projecting to the Sm (39.7%) was significantly higher than that projecting to the LH (5.4%) or VPM (5.6%; P<.001). vpm 232-235 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 12949783-8 2003 In the caudal ventrolateral medulla and nucleus tractus solitarius, Fos-positive neurons projected to the Sm, PB, and LH. Samarium 106-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 14559159-0 2003 MK-801 attenuates cocaine induction of c-fos and preprodynorphin mRNA levels in Fischer rats. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 14559159-0 2003 MK-801 attenuates cocaine induction of c-fos and preprodynorphin mRNA levels in Fischer rats. Cocaine 18-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 14559159-1 2003 This study shows that single (15 mg/kg) or binge (3x15 mg/kg) cocaine administration increases c-fos mRNA levels, but only binge cocaine administration increases preprodynorphin mRNA levels in the caudate/putamen of Fischer rats. Cocaine 62-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 14559159-2 2003 This increase in both c-fos and preprodynorphin mRNA levels is attenuated by the noncompetitive NMDA antagonist MK-801 (0.25 mg/kg), suggesting a preferential role for NMDA receptor co-activation in cellular events leading to cocaine-induced behaviors. N-Methylaspartate 96-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 14559159-2 2003 This increase in both c-fos and preprodynorphin mRNA levels is attenuated by the noncompetitive NMDA antagonist MK-801 (0.25 mg/kg), suggesting a preferential role for NMDA receptor co-activation in cellular events leading to cocaine-induced behaviors. Dizocilpine Maleate 112-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 14559159-2 2003 This increase in both c-fos and preprodynorphin mRNA levels is attenuated by the noncompetitive NMDA antagonist MK-801 (0.25 mg/kg), suggesting a preferential role for NMDA receptor co-activation in cellular events leading to cocaine-induced behaviors. Cocaine 226-233 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 12965227-5 2003 The results show that intrastriatal amphetamine increases wakefulness independent of motor activity, and it increases c-Fos expression in the PPT and adjacent areas. Amphetamine 36-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 14574230-2 2003 Recently, we found that nuclear RACK1 mediates acute ethanol induction of immediate early gene c-fos expression. Ethanol 53-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 14574230-14 2003 However, transduction of exogenous RACK1 expressed as a Tat-fusion protein was able to rescue c-fos mRNA expression after chronic ethanol exposure. Ethanol 130-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 14574230-15 2003 CONCLUSIONS: Our data suggest that RACK1 nuclear compartmentalization and ethanol-induced c-fos expression are transient and are desensitized to ethanol during prolonged exposure to high concentrations. Ethanol 74-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 14574230-17 2003 Our data further suggest that the altered compartmentalization of RACK1 leads to differences in c-fos expression upon acute or chronic exposure to ethanol. Ethanol 147-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 14574230-18 2003 In summary, RACK1 is an important molecular mediator of the acute and chronic actions of ethanol on the expression of c-fos. Ethanol 89-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 14622196-5 2003 injection of formalin as a chemical somatic noxious stimulus increased c-fos mRNA expression in the BLA, but not CeA, while intraperitoneal (i.p.) Formaldehyde 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 14553905-8 2003 In animals with bilateral cochlear ablation the number of c-Fos reactive cell nuclei representing basal expression was generally low in the VCN and DCN of both sides. dcn 148-151 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 13130519-0 2003 Suppressive effects of BmK IT2 on nociceptive behavior and c-Fos expression in spinal cord induced by formalin. Formaldehyde 102-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 13130519-3 2003 The detection of c-Fos expression induced by formalin in either the absence or the presence of BmK IT2 was carried out with the ABC method. Formaldehyde 45-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 13130519-7 2003 In addition, c-Fos expression induced by formalin was significantly inhibited in all laminae of L4-5 spinal cord by pre- or posttreatment with BmK IT2. Formaldehyde 41-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 13130519-9 2003 In addition, the nonparallel suppression of BmK IT2 on flinch behavior and c-Fos expression induced by formalin may be ascribed to the different activity patterns of afferent fibers and central neurons. Formaldehyde 103-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 12941373-6 2003 This anti-nociceptive effect was accompanied by an HU210-induced attenuation of the formalin-evoked increase in Fos protein expression in the caudal lateral PAG. HU 211 51-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 12941373-6 2003 This anti-nociceptive effect was accompanied by an HU210-induced attenuation of the formalin-evoked increase in Fos protein expression in the caudal lateral PAG. Formaldehyde 84-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 12900043-2 2003 A previous Fos study also revealed a marked activation of the ventrolateral part of the periaqueductal gray (VLPAG) and a much smaller activation of its dorsal part (DPAG). dpag 166-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 14508329-9 2003 RESULTS: Cotransfection of c-Fos antibody significantly decreased glutamate-induced AP-1 activity. Glutamic Acid 66-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 14508330-5 2003 RESULTS: Microinjection of either naloxone (an opioid receptor antagonist) or muscimol (a gamma-aminobutyric acid receptor type A agonist) into the ventrolateral periaqueductal gray area inhibited N2O-induced c-Fos expression in the spinal cord and pontine noradrenergic nuclei, particularly in the A7. Naloxone 34-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 209-214 14508330-5 2003 RESULTS: Microinjection of either naloxone (an opioid receptor antagonist) or muscimol (a gamma-aminobutyric acid receptor type A agonist) into the ventrolateral periaqueductal gray area inhibited N2O-induced c-Fos expression in the spinal cord and pontine noradrenergic nuclei, particularly in the A7. Muscimol 78-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 209-214 14508330-6 2003 Microinjection of either naloxone or muscimol into the A7 nuclei also inhibited N2O-induced c-Fos expression in the spinal cord and the N2O-induced antinociceptive effect by the plantar test. Naloxone 25-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 14508330-6 2003 Microinjection of either naloxone or muscimol into the A7 nuclei also inhibited N2O-induced c-Fos expression in the spinal cord and the N2O-induced antinociceptive effect by the plantar test. Muscimol 37-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 14508330-6 2003 Microinjection of either naloxone or muscimol into the A7 nuclei also inhibited N2O-induced c-Fos expression in the spinal cord and the N2O-induced antinociceptive effect by the plantar test. Nitrous Oxide 80-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 14619585-6 2003 CONCLUSION: Intrathecal neostigmine could induce nitric oxide release in the spinal cord, the suppression of Fos expression might be one of the antinociception mechanisms of intrathecal neostigmine in the formalin test. Neostigmine 186-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-112 14619585-6 2003 CONCLUSION: Intrathecal neostigmine could induce nitric oxide release in the spinal cord, the suppression of Fos expression might be one of the antinociception mechanisms of intrathecal neostigmine in the formalin test. Formaldehyde 205-213 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-112 14554105-4 2003 In addition, the increase of c-Fos expression in laminae I-II, V-VI, and VII-X induced by BmK I, and not in laminae III-IV, could be partially inhibited by systemic morphine in a dose-dependent manner. Morphine 165-173 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 12963647-7 2003 Longitudinal transcriptional profiling revealed that simvastatin downregulated the inflammatory genes fos, jun, and tumor necrosis factor-alpha and upregulated the cell cycle inhibitor p27Kip1, endothelial nitric oxide synthase, and bone morphogenetic protein receptor type 1a. Simvastatin 53-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 13679249-8 2003 Repeated injections of carbachol (30 pmol) into the LSD produced Fos immunoreactivity in the ipsilateral side of the LSV. Carbachol 23-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 14766052-8 2003 However, following treatment with H7 inhibited the expression of c-fos mRNA and protein, MMP-1,2/TIMP-1,2 proteins expression decreased. 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine 34-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 12827644-7 2003 In addition, intermittent L-DOPA induced an increase in the mRNA levels encoding for the 65 kDa isoform of glutamate decarboxylase in globus pallidus neurons ipsilateral to the lesion and a bilateral increase in c-fos mRNA expression in the subthalamic nucleus. Levodopa 26-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 212-217 14601197-2 2003 METHODS: The antinociceptive effects were assessed in male Sprague-Dawley rats by behavioral responses to phasic and tonic nociceptive stimulations and biochemical index of pain, formalin-induced Fos-like immunoreactivity (Fos-LI), in spinal cord dorsal horn. Formaldehyde 179-187 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 196-199 14601197-2 2003 METHODS: The antinociceptive effects were assessed in male Sprague-Dawley rats by behavioral responses to phasic and tonic nociceptive stimulations and biochemical index of pain, formalin-induced Fos-like immunoreactivity (Fos-LI), in spinal cord dorsal horn. Formaldehyde 179-187 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 223-226 14601197-9 2003 Fentanyl increased the threshold level to noxious thermal stimulation, and reduced the formalin-induced licking/biting behaviors and the number of Fos-LI labelled neurons which are predominantly found in the neck of the dorsal horn. Fentanyl 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-150 12960557-6 2003 RESULTS: Inhalation of nitrous oxide induced a dose-dependent increase in c-Fos expression in CRF-positive neurons in the paraventricular nucleus of the hypothalamus. Nitrous Oxide 23-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 12960557-7 2003 Intracerebroventricular administration of CRF antagonist inhibited nitrous oxide-induced c-Fos expression in the locus ceruleus and the antinociceptive effect of nitrous oxide. Nitrous Oxide 67-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 12956728-0 2003 c-Fos and peptide immunoreactivities in the central extended amygdala of morphine-dependent rats after naloxone-precipitated withdrawal. Morphine 73-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 12956728-0 2003 c-Fos and peptide immunoreactivities in the central extended amygdala of morphine-dependent rats after naloxone-precipitated withdrawal. Naloxone 103-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 12956728-3 2003 Naloxone treatment in naive rats induced a slight increase in c-Fos immunoreactivity in the central amygdaloid nucleus, the lateral bed nucleus of the stria terminalis and the interstitial nucleus of the posterior limb of the anterior commissure. Naloxone 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 12956728-4 2003 In morphine-dependent rats, naloxone injection significantly increased the number of c-Fos-positive neurons in these structures as well as in the majority of the other central extended amygdala components. Morphine 3-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 12956728-4 2003 In morphine-dependent rats, naloxone injection significantly increased the number of c-Fos-positive neurons in these structures as well as in the majority of the other central extended amygdala components. Naloxone 28-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 12956728-6 2003 Corticotropin-releasing factor- and methionine-enkephakin-immunoreactive neurons displayed c-Fos immunoreactivity in naive rats after naloxone injection, whereas only enkephalinergic neurons were found to be c-Fos positive in morphine-dependent rats after naloxone injection. Naloxone 134-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 14511337-5 2003 Using in situ hybridization histochemistry, we assessed the effects of acute and repeated methylphenidate treatment on the expression of immediate-early genes (c-fos, zif 268) and neuropeptides (dynorphin, substance P, enkephalin) in adolescent rats. Methylphenidate 90-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 14511337-10 2003 Moreover, after repeated methylphenidate treatment, cocaine-induced expression of c-fos and zif 268, as well as of substance P, was significantly attenuated throughout the striatum. Methylphenidate 25-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-99 14511337-10 2003 Moreover, after repeated methylphenidate treatment, cocaine-induced expression of c-fos and zif 268, as well as of substance P, was significantly attenuated throughout the striatum. Cocaine 52-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-99 12967638-7 2003 Expression of c-fos mRNA and rates of protein synthesis were measured by northern blot analysis and by 14C-phenylalanine incorporation, respectively. 14c-phenylalanine 103-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 12967638-9 2003 Cyclosporin A and FK506, the calcineurin inhibitors, significantly inhibited the increases in both c-fos expression and protein synthesis. Cyclosporine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 12967638-9 2003 Cyclosporin A and FK506, the calcineurin inhibitors, significantly inhibited the increases in both c-fos expression and protein synthesis. Tacrolimus 18-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 14613087-11 2003 Morphine dose dependently attenuates the number of behaviors, as well as Fos expression; this effect is reversed by the micro-opioid receptor antagonist naltrexone. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-76 14613087-11 2003 Morphine dose dependently attenuates the number of behaviors, as well as Fos expression; this effect is reversed by the micro-opioid receptor antagonist naltrexone. Naltrexone 153-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-76 12900043-4 2003 We now test the role of the DPAG to see if its small activation (as revealed by Fos) is of any functional significance in the contextual fear response. dpag 28-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 12842125-0 2003 Bilateral lesions of the entorhinal cortex differentially modify haloperidol- and olanzapine-induced c-fos mRNA expression in the rat forebrain. Haloperidol 65-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 12873950-0 2003 The effects of pyrilamine and cimetidine on mRNA C-fos expression and nociceptive flinching behavior in rats. Pyrilamine 15-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 12873950-0 2003 The effects of pyrilamine and cimetidine on mRNA C-fos expression and nociceptive flinching behavior in rats. Cimetidine 30-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 12873950-8 2003 Pyrilamine 5 and 20 mM decreased c-fos mRNA expression, and cimetidine decreased the expression only at 100 mM. Pyrilamine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 12928072-0 2003 c-Fos immunoreactivity in the brain after esophageal acid stimulation. esophageal acid 42-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 12834432-6 2003 Of the neurones in the subfornical organ that were shown by retrograde labelling to project to BNST, approximately 50% expressed Fos in response to isoproterenol. Isoproterenol 148-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-132 12842125-0 2003 Bilateral lesions of the entorhinal cortex differentially modify haloperidol- and olanzapine-induced c-fos mRNA expression in the rat forebrain. Olanzapine 82-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 12842125-3 2003 After a 15-day recovery period, the effect of a typical antipsychotic, haloperidol (1 mg/kg), on c-fos mRNA expression was compared with that of an atypical one, olanzapine (10 mg/kg), in both sham-lesioned and entorhinal cortex-lesioned rats. Haloperidol 71-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 12842125-4 2003 In sham-lesioned rats, both haloperidol and olanzapine induced c-fos expression in the caudal cingulate cortex, dorsomedial and dorsolateral caudate-putamen, nucleus accumbens core and shell and lateral septum. Haloperidol 28-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 12842125-4 2003 In sham-lesioned rats, both haloperidol and olanzapine induced c-fos expression in the caudal cingulate cortex, dorsomedial and dorsolateral caudate-putamen, nucleus accumbens core and shell and lateral septum. Olanzapine 44-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 12842125-5 2003 In addition, olanzapine, but not haloperidol, increased c-fos expression within the central amygdala. Olanzapine 13-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 12842125-6 2003 In entorhinal cortex-lesioned rats, haloperidol-induced c-fos expression was markedly reduced in most areas. Haloperidol 36-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 12842125-7 2003 In contrast, the olanzapine-induced c-fos expression was not altered in the nucleus accumbens shell and lateral septum of the lesioned rats. Olanzapine 17-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 12834907-0 2003 Electroacupuncture induces the expression of Fos in rat dorsal horn via capsaicin-insensitive afferents. Capsaicin 72-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 12812860-7 2003 Intravascular administration of apomorphine had no effect on basal ACTH levels but did substantially increase the number of Fos-positive amygdala and nucleus tractus solitarius catecholamine cells. Apomorphine 32-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-127 12812860-8 2003 Administration of apomorphine prior to immune challenge augmented the normal ACTH response to this stressor at 90 min and there was a corresponding increase in the number of Fos-positive paraventricular nucleus corticotropin-releasing factor cells, paraventricular nucleus oxytocin cells and nucleus tractus solitarius catecholamine cells. Apomorphine 18-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 174-177 12842287-5 2003 The number of Fos-positive neurons following intestinal anaphylaxis decreased in animals exposed to water-avoidance stress (P < 0.05), although serum levels of rat mast cell protease II were not different in stressed and unstressed animals, indicating a similar degree of mast cell degranulation. Water 100-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 12821175-4 2003 As expected, a significant induction of the immediate early gene Fos was observed after acute administration of morphine, cocaine, 3, 4-methylenedioxymethamphetamine and Delta(9)-Tetrahydrocannabinol. Morphine 112-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 12821175-4 2003 As expected, a significant induction of the immediate early gene Fos was observed after acute administration of morphine, cocaine, 3, 4-methylenedioxymethamphetamine and Delta(9)-Tetrahydrocannabinol. Cocaine 122-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 12834907-4 2003 Fos expression in the dorsal horn after injection of formalin into the hindpaw was severely attenuated by neonatal capsaicin treatment. Formaldehyde 53-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 12821175-4 2003 As expected, a significant induction of the immediate early gene Fos was observed after acute administration of morphine, cocaine, 3, 4-methylenedioxymethamphetamine and Delta(9)-Tetrahydrocannabinol. N-Methyl-3,4-methylenedioxyamphetamine 131-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 12821175-4 2003 As expected, a significant induction of the immediate early gene Fos was observed after acute administration of morphine, cocaine, 3, 4-methylenedioxymethamphetamine and Delta(9)-Tetrahydrocannabinol. Dronabinol 170-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 12834907-4 2003 Fos expression in the dorsal horn after injection of formalin into the hindpaw was severely attenuated by neonatal capsaicin treatment. Capsaicin 115-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 12834907-6 2003 These results suggest that E-acupuncture induces the expression of Fos in the dorsal horn neurons via capsaicin-insensitive afferents, presumably Adelta afferents. Capsaicin 102-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 12831996-5 2003 Four hours after carrageenan injection, in the LC/SC both ipsilateral and contralateral to the inflamed paw, the number of Fos-positive cells increased significantly in carrageenan-injected rats when compared to vehicle (saline)-injected and untreated control rats. Carrageenan 17-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-126 12831996-5 2003 Four hours after carrageenan injection, in the LC/SC both ipsilateral and contralateral to the inflamed paw, the number of Fos-positive cells increased significantly in carrageenan-injected rats when compared to vehicle (saline)-injected and untreated control rats. Carrageenan 169-180 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-126 12831996-6 2003 The Fos expression in the LC/SC was equivalent bilaterally in the carrageenan-injected rats, as well as in vehicle-injected and untreated control rats. Carrageenan 66-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-7 12821377-3 2003 Both cocaine binge (3 x 15 mg/kg) and perinatal asphyxia on embryonic day 22 (E22) induced c-fos in the striatum as well as in several other brain regions within 3 h after treatment. Cocaine 5-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 12865328-6 2003 Pretreatment with corticosterone decreased the number of Fos-immunoreactive cells in the PVN and SON but not in the DMH. Corticosterone 18-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 12853419-0 2003 c-fos reduces corticosterone-mediated effects on neurotrophic factor expression in the rat hippocampal CA1 region. Corticosterone 14-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 12853419-5 2003 c-fos antisense oligodeoxynucleotides were injected into the dorsal hippocampus of adrenalectomized rats. Oligodeoxyribonucleotides 16-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 12812839-4 2003 We found that intraplantar injection of formalin as a chemical somatic noxious stimulus increased c-fos mRNA expression in the La and BL, but not Ce. Formaldehyde 40-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 12826861-10 2003 Uterine cervical distension-evoked cFos expression was prevented by prior infiltration of lidocaine into the cervix. Lidocaine 90-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-39 12826861-11 2003 Intrathecal ketorolac produced a dose-dependent inhibition of uterine cervical distension-induced cFos expression. Ketorolac 12-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-102 12826861-13 2003 The inhibition of cFos expression by intrathecal ketorolac suggests that spinal cyclo-oxygenase plays a role in uterine cervical distension-induced nociception. Ketorolac 49-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-22 12821377-4 2003 Maternal administration of a D1 dopamine antagonist, SCH 23390, before either cocaine or asphyxia exposure dramatically reduced the numbers of Fos-immunoreactive cells in the striatum as well as in many other brain regions. Dopamine 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-146 12788208-0 2003 Fos induction in the rat deep cerebellar and vestibular nuclei following central administration of colchicine: a qualitative and quantitative time-course study. Colchicine 99-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 12887411-8 2003 Coadministration of CGS 21680 and CPA produced a synergistic induction of c-fos expression in the caudate-putamen, cingulate cortex, and especially the NAc. cysteinylglycine 20-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 12887411-10 2003 Because it has been previously shown that activation of dopamine D2 receptors (D2Rs) by endogenous dopamine blocks A2AR-mediated c-fos expression, it is hypothesized that the enabling role of A1Rs in A2AR-mediated striatal c-fos expression is related to the A1R-mediated inhibition of dopamine release. Dopamine 56-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 12887411-10 2003 Because it has been previously shown that activation of dopamine D2 receptors (D2Rs) by endogenous dopamine blocks A2AR-mediated c-fos expression, it is hypothesized that the enabling role of A1Rs in A2AR-mediated striatal c-fos expression is related to the A1R-mediated inhibition of dopamine release. Dopamine 99-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 12855312-6 2003 In intact rats, c-Fos expression in the paraventricular nucleus of the thalamus and arcuate nucleus of the hypothalamus remained at control levels or decreased in animals injected two or three times with formalin; in gonadectomized rats, c-Fos expression increased with repetition of the noxious stimulation, reaching the highest levels in animals injected three times with formalin. Formaldehyde 204-212 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 12855312-6 2003 In intact rats, c-Fos expression in the paraventricular nucleus of the thalamus and arcuate nucleus of the hypothalamus remained at control levels or decreased in animals injected two or three times with formalin; in gonadectomized rats, c-Fos expression increased with repetition of the noxious stimulation, reaching the highest levels in animals injected three times with formalin. Formaldehyde 204-212 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 238-243 12855312-6 2003 In intact rats, c-Fos expression in the paraventricular nucleus of the thalamus and arcuate nucleus of the hypothalamus remained at control levels or decreased in animals injected two or three times with formalin; in gonadectomized rats, c-Fos expression increased with repetition of the noxious stimulation, reaching the highest levels in animals injected three times with formalin. Formaldehyde 374-382 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 12910709-0 2003 [Effect of Fos expression induced by spinal nitric oxide on the nociception of formalin in rats]. Nitric Oxide 44-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 12910709-0 2003 [Effect of Fos expression induced by spinal nitric oxide on the nociception of formalin in rats]. Formaldehyde 79-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 12810563-6 2003 These findings indicate an inhibitory role for testosterone on stress-induced indexes of synaptic (Fos) and transcriptional (AVP hnRNA) activation among hypophysiotropic paraventricular neurons and provide meaningful end points with which to pursue how and where androgens operate on stress-related input to the paraventricular nucleus motor neurons. Testosterone 47-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 12788208-4 2003 On the other hand, moderate number of Fos-positive cells was visible in each of the cerebellar and vestibular nuclei 24h after colchicine treatment. Colchicine 127-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 12788208-5 2003 Exposure of the animals to 48h of colchicine treatment induced an additional, more than 50%, rise in the accumulation of Fos-positive profiles in almost all the cerebellar and vestibular nuclei. Colchicine 34-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-124 12788208-1 2003 The present study was conducted to demonstrate Fos expression at four levels (anterior, prefastigial, postfastigial, posterior) of the cerebellar-vestibular nuclear complex in rats exposed to 1, 6, 24, and 48h of colchicine treatment using a light microscopic avidin biotin peroxidase (ABC) immunohistochemistry. Colchicine 213-223 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 12788208-8 2003 We assume that the delayed Fos activation in these structures indicate that the cerebellar and vestibular nuclei are not the primary targets of the central effect of colchicine and their activation seems to be rather a result of a postponed functional consequences of the central action of colchicine probably related to the coordination of motor performance. Colchicine 166-176 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 12930673-0 2003 [Expression of proto-oncogenes c-fos and c-jun in osteoblasts activated by excessive fluoride]. Fluorides 85-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 12930673-1 2003 OBJECTIVE: To study expression of proto-oncogenes c-fos and its accompanying gene c-jun in osteoblasts activated by action of excessive fluoride in vivo and in vitro. Fluorides 136-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 12930673-4 2003 RESULTS: Sodium fluoride could stimulate the proliferation of osteoblast in rats with chronic fluorosis and induce expression of both c-fos and c-jun in all envelops of the spine bone, as compared with its control group. Sodium Fluoride 9-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-139 12930673-5 2003 Value of optical absorption in mRNA expression of c-fos and c-jun was 139.63 and 126.37, respectively, in rats with NaF plus high-calcium, significantly lower than that in control group with high-calcium only (107.74 and 117.48, respectively) (P < 0.001). Sodium Fluoride 116-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 12930673-5 2003 Value of optical absorption in mRNA expression of c-fos and c-jun was 139.63 and 126.37, respectively, in rats with NaF plus high-calcium, significantly lower than that in control group with high-calcium only (107.74 and 117.48, respectively) (P < 0.001). Calcium 130-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 12930673-5 2003 Value of optical absorption in mRNA expression of c-fos and c-jun was 139.63 and 126.37, respectively, in rats with NaF plus high-calcium, significantly lower than that in control group with high-calcium only (107.74 and 117.48, respectively) (P < 0.001). Calcium 196-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 12930673-7 2003 Western blotting showed that level of protein expression of c-fos and c-jun in periosteal osteoblasts was significantly higher in all rat groups with NaF than that in all control groups, with values of optical absorption of 123.32, 116.60, 115.97 and 108.30, respectively. Sodium Fluoride 150-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 12930673-8 2003 mRNA expression of c-fos and c-jun in osteoblast-like cells treated with NaF for 12 h increased obviously, and remained at high level 48 h after exposure, with values of optical absorption of 114.80, 161.14, 118.20, and 150.41, respectively, as compared with that in control group (P < 0.001 and P < 0.05). Sodium Fluoride 73-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 12930673-9 2003 CONCLUSIONS: Exposure to excessive fluoride could stimulate activation and proliferation of both osteoblasts in rats and cultured osteoblast-like cells in vitro, and cause enhanced expression of mRNA and protein of both c-fos and c-jun. Fluorides 35-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 220-225 12810678-13 2003 Finally, the cytotoxicity and radiosensitizing effects of 2DG were more pronounced in v-Fos-transformed versus nontransformed immortalized rat cells, and this radiosensitization was also inhibited by treatment with NAC. Deoxyglucose 58-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 12810678-13 2003 Finally, the cytotoxicity and radiosensitizing effects of 2DG were more pronounced in v-Fos-transformed versus nontransformed immortalized rat cells, and this radiosensitization was also inhibited by treatment with NAC. Acetylcysteine 215-218 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 12759135-1 2003 In this study, we evaluated the effects of intrathecally administered agonists of mu- and delta-opioid receptor and their analogs on the pain-induced behavior and expression of c-Fos immunoreactivity in the spinal cord, elicited by intraplantar injection of 12% formalin to the hindpaw of the rat. Formaldehyde 262-270 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 177-182 12759135-3 2003 In this study, intrathecal injection of morphine (10 microg), endomorphin-2 (5 microg) and its analog Dmt-endomorphin-2 (10 microg) significantly decreased the formalin-induced pain behavior, and lowered a number of c-Fos positive neurons in the laminae I, II and III of the spinal cord by about 40%, 30% and 40%, respectively. Morphine 40-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 216-221 12759135-3 2003 In this study, intrathecal injection of morphine (10 microg), endomorphin-2 (5 microg) and its analog Dmt-endomorphin-2 (10 microg) significantly decreased the formalin-induced pain behavior, and lowered a number of c-Fos positive neurons in the laminae I, II and III of the spinal cord by about 40%, 30% and 40%, respectively. dmt 102-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 216-221 12759135-3 2003 In this study, intrathecal injection of morphine (10 microg), endomorphin-2 (5 microg) and its analog Dmt-endomorphin-2 (10 microg) significantly decreased the formalin-induced pain behavior, and lowered a number of c-Fos positive neurons in the laminae I, II and III of the spinal cord by about 40%, 30% and 40%, respectively. endomorphin 2 62-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 216-221 12759135-3 2003 In this study, intrathecal injection of morphine (10 microg), endomorphin-2 (5 microg) and its analog Dmt-endomorphin-2 (10 microg) significantly decreased the formalin-induced pain behavior, and lowered a number of c-Fos positive neurons in the laminae I, II and III of the spinal cord by about 40%, 30% and 40%, respectively. Formaldehyde 160-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 216-221 12854134-8 2003 The OD value of c-fos protein in hydrotalcite group was higher than that in ranitidine group and control group (0.52+/-0.07 vs 0.31+/-0.04, 0.32+/-0.05, P<0.05). Ranitidine 76-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 12854134-9 2003 CONCLUSION: Hydrotalcite can protect gastric mucosal injury in rats induced by taurocholate, which may be related to the increased expression of TFF2 and c-fos protein. Taurocholic Acid 79-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 15139112-3 2003 RESULT: Parthenolide inhibited protein levels of c-fos, c-myc in a time-dependent manner but didn"t affect the protein levels of p15, p16, p18, p19. parthenolide 8-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 15139112-6 2003 CONCLUSION: Parthenolide may inhibit proliferation of VSMC by inhibiting the expressions of c-fos, c-myc, but not the expressions of p15, p16, p18, p19. parthenolide 12-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 15139112-6 2003 CONCLUSION: Parthenolide may inhibit proliferation of VSMC by inhibiting the expressions of c-fos, c-myc, but not the expressions of p15, p16, p18, p19. vsmc 54-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 12801600-6 2003 In both NT knockout mice and rats pretreated with SR 48692, haloperidol-induced Fos expression was markedly attenuated in the dorsolateral striatum; amphetamine-induced Fos expression was reduced in the medial striatum. SR 48692 50-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 12801600-6 2003 In both NT knockout mice and rats pretreated with SR 48692, haloperidol-induced Fos expression was markedly attenuated in the dorsolateral striatum; amphetamine-induced Fos expression was reduced in the medial striatum. SR 48692 50-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-172 12801600-6 2003 In both NT knockout mice and rats pretreated with SR 48692, haloperidol-induced Fos expression was markedly attenuated in the dorsolateral striatum; amphetamine-induced Fos expression was reduced in the medial striatum. Haloperidol 60-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 12801600-6 2003 In both NT knockout mice and rats pretreated with SR 48692, haloperidol-induced Fos expression was markedly attenuated in the dorsolateral striatum; amphetamine-induced Fos expression was reduced in the medial striatum. Amphetamine 149-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-172 12809946-7 2003 These results suggested that selenite induced apoptosis accompanied by the activation of caspase-3 and JNK and the upregulation of c-jun, c-fos, p53 and p21(WAF1/CIP1) at the early stage of liver regeneration. Selenious Acid 29-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 12717712-0 2003 Fos expression by glutamatergic neurons of the solitary tract nucleus after phenylephrine-induced hypertension in rats. Phenylephrine 76-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 12717712-6 2003 Saline-treated rats had very few Fos-ir neurons. Sodium Chloride 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 12742625-12 2003 Urocortin induced a similar pattern of Fos response in the brain and the spinal cord. Urocortins 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 12742633-3 2003 Both electrical stimulation and chemical (0.3 mM bicuculline methobromide) activation of the dorsal PAG elicited a selective increase in Fos-like immunoreactivity (FLI) in the superior lateral and central lateral subnuclei of the rostral lateral PBN (LPBN) relative to surgery and blood pressure control groups. bicuculline methobromide 49-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-140 12742639-10 2003 Similar to opioids counteracting noxiously induced c-Fos expression, 10 mg/kg (s.c.) of morphine reduced the number of c-Fos-IR nuclei induced by 0.63 mg/kg of F 13640 (by 45+/-5%; P<0.001). Morphine 88-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 12742639-10 2003 Similar to opioids counteracting noxiously induced c-Fos expression, 10 mg/kg (s.c.) of morphine reduced the number of c-Fos-IR nuclei induced by 0.63 mg/kg of F 13640 (by 45+/-5%; P<0.001). Morphine 88-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 12788208-2 2003 Intracerebroventricular administration of colchicine (60microg per 10microl saline) elicited a continuous increase in the number of Fos-positive cells in the main cerebellar (fastigial, interpositus, dentatus) and vestibular (superior, medial, lateral, spinal, Y) nuclei. Colchicine 42-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-135 12788208-3 2003 One and six hours after colchicine treatment, intensive Fos labeling was observed only in the pyriform cortex and the hypothalamic paraventricular nucleus, respectively, and there was no Fos immunolabeling in any of the cerebellar or vestibular structures investigated. Colchicine 24-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 12736179-4 2003 In the perifornical area, a significantly higher percentage of orexin neurons expressed Fos after muscimol compared with saline injection. Muscimol 98-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 12948621-7 2003 The increase of c-fos expression in time correlated best with the increase of glutamate release, which was also significantly elevated at every sampling time. Glutamic Acid 78-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 12948621-10 2003 We conclude that glutamate plays the essential role (most probably through ionotropic and metabotropic receptors) in the extracellular signaling, which eventually leads to intracellular cascades and c-fos gene expression in the striatum during convulsions. Glutamic Acid 17-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-204 12787062-0 2003 c-Fos is essential for the response of the tyrosine hydroxylase gene to depolarization or phorbol ester. Phorbol Esters 90-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 12814374-0 2003 Morphine withdrawal precipitated by specific mu, delta or kappa opioid receptor antagonists: a c-Fos protein study in the rat central nervous system. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 12814374-1 2003 We have recently shown concurrent changes in behavioural responses and c-Fos protein expression in the central nervous system in both naive and morphine-dependent rats after systemic administration of the opioid antagonist naloxone. Morphine 144-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 12814374-1 2003 We have recently shown concurrent changes in behavioural responses and c-Fos protein expression in the central nervous system in both naive and morphine-dependent rats after systemic administration of the opioid antagonist naloxone. Naloxone 223-231 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 12814374-3 2003 In a first experimental series, only beta-funaltrexamine increased c-Fos expression in the eight central nervous system structures examined, whereas no effect was seen after naltrindole or nor-binaltorphimine administration in naive rats. beta-funaltrexamine 37-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 12814374-5 2003 A second experimental series in morphine-dependent rats showed that beta-funaltrexamine had the highest potency in the induction of classical signs of morphine withdrawal syndrome, as well as the increase in c-Fos expression in the 22 central nervous system structures studied, suggesting a major role of mu opioid receptors in opioid dependence. Morphine 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 208-213 12814374-6 2003 However, our results also demonstrated that naltrindole and, to a lesser extent, nor-binaltorphimine were able to induce moderate signs of morphine withdrawal and relatively weak c-Fos protein expression in restricted central nervous system structures. naltrindole 44-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-184 12814374-6 2003 However, our results also demonstrated that naltrindole and, to a lesser extent, nor-binaltorphimine were able to induce moderate signs of morphine withdrawal and relatively weak c-Fos protein expression in restricted central nervous system structures. norbinaltorphimine 81-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-184 12787062-8 2003 Stimulation of TH gene promoter activity, which was observed in control cell lines treated with either 50 mm KCl or TPA was also dramatically inhibited in the c-Fos-deficient cells. Potassium Chloride 109-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-164 12787062-8 2003 Stimulation of TH gene promoter activity, which was observed in control cell lines treated with either 50 mm KCl or TPA was also dramatically inhibited in the c-Fos-deficient cells. Tetradecanoylphorbol Acetate 116-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-164 12727327-0 2003 Fos protein expression following acute administration of diethyldithiocarbamate in rats. Ditiocarb 57-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 12818716-4 2003 In the present study, we tested whether ETOH[ICV] could induce a conditioned taste preference (CTP) or aversion (CTA) and alter c-Fos immunoreactivity (c-Fos-IR) in brain regions associated with feeding, aversion, and/or reward. Ethanol 40-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-133 12818716-4 2003 In the present study, we tested whether ETOH[ICV] could induce a conditioned taste preference (CTP) or aversion (CTA) and alter c-Fos immunoreactivity (c-Fos-IR) in brain regions associated with feeding, aversion, and/or reward. Ethanol 40-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-157 12818716-4 2003 In the present study, we tested whether ETOH[ICV] could induce a conditioned taste preference (CTP) or aversion (CTA) and alter c-Fos immunoreactivity (c-Fos-IR) in brain regions associated with feeding, aversion, and/or reward. icv 45-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-133 12818716-4 2003 In the present study, we tested whether ETOH[ICV] could induce a conditioned taste preference (CTP) or aversion (CTA) and alter c-Fos immunoreactivity (c-Fos-IR) in brain regions associated with feeding, aversion, and/or reward. icv 45-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-157 12818716-8 2003 ETOH[ICV] significantly increased c-Fos-IR in a number of brain sites associated with feeding and reward including the bed nucleus of the stria terminalis, lateral dorsal area (BSTLD); nucleus accumbens, shell area (AcbSh); hypothalamic paraventricular nucleus (PVN); and lateral septum, ventral area (LSV). Ethanol 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 12818716-9 2003 Thus, ETOH induced a CTP, not CTA, via central mechanisms; it increased c-Fos-IR in specific sites associated with feeding and reward. Ethanol 6-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 12711372-1 2003 We investigated the ability of dextromethorphan, a clinically available NMDA receptor antagonist, to attenuate the behaviors and the expression of c-fos mRNA associated with acute morphine withdrawal in the 7-day-old rat. Dextromethorphan 31-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-152 12711372-1 2003 We investigated the ability of dextromethorphan, a clinically available NMDA receptor antagonist, to attenuate the behaviors and the expression of c-fos mRNA associated with acute morphine withdrawal in the 7-day-old rat. Morphine 180-188 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-152 12711372-2 2003 The intensity of the acute morphine withdrawal behaviors and the elevation in c-fos mRNA expression in the brain induced by acute morphine withdrawal were reduced by dextromethorphan. Morphine 130-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 12711372-2 2003 The intensity of the acute morphine withdrawal behaviors and the elevation in c-fos mRNA expression in the brain induced by acute morphine withdrawal were reduced by dextromethorphan. Dextromethorphan 166-182 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 12767478-1 2003 In this study, we examined the effect of the s.c. administration of (+/-) 3,4-methylenedioxymethamphetamine (MDMA) or saline on locomotor activity and Fos expression following the bilateral destruction of hippocampal dentate granule cells by colchicine in rats. N-Methyl-3,4-methylenedioxyamphetamine 68-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-154 12767478-1 2003 In this study, we examined the effect of the s.c. administration of (+/-) 3,4-methylenedioxymethamphetamine (MDMA) or saline on locomotor activity and Fos expression following the bilateral destruction of hippocampal dentate granule cells by colchicine in rats. N-Methyl-3,4-methylenedioxyamphetamine 109-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-154 12767478-1 2003 In this study, we examined the effect of the s.c. administration of (+/-) 3,4-methylenedioxymethamphetamine (MDMA) or saline on locomotor activity and Fos expression following the bilateral destruction of hippocampal dentate granule cells by colchicine in rats. Sodium Chloride 118-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-154 12767478-3 2003 In addition, when the lesioned animals were given s.c. saline or MDMA, there was a significant increase in Fos expression in the nucleus accumbens core, but not in the medial prefrontal cortex, dorsolateral prefrontal cortex, anterior cingulate cortex, piriform cortex, dorsal striatum, or nucleus accumbens shell, compared to the intact and sham-lesioned animals. Sodium Chloride 55-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 12767478-3 2003 In addition, when the lesioned animals were given s.c. saline or MDMA, there was a significant increase in Fos expression in the nucleus accumbens core, but not in the medial prefrontal cortex, dorsolateral prefrontal cortex, anterior cingulate cortex, piriform cortex, dorsal striatum, or nucleus accumbens shell, compared to the intact and sham-lesioned animals. N-Methyl-3,4-methylenedioxyamphetamine 65-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 12709776-2 2003 OBJECTIVE: For this study, the pattern of expression of the protein Fos, a marker of cellular activation, was compared after administration of the typical neuroleptic haloperidol, the antipsychotic drug clozapine, and the atypical neuroleptic olanzapine, as well as the sedative drug diphenhydramine and the anxiolytic lorazepam. Haloperidol 167-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 12709776-2 2003 OBJECTIVE: For this study, the pattern of expression of the protein Fos, a marker of cellular activation, was compared after administration of the typical neuroleptic haloperidol, the antipsychotic drug clozapine, and the atypical neuroleptic olanzapine, as well as the sedative drug diphenhydramine and the anxiolytic lorazepam. Olanzapine 243-253 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 12709776-2 2003 OBJECTIVE: For this study, the pattern of expression of the protein Fos, a marker of cellular activation, was compared after administration of the typical neuroleptic haloperidol, the antipsychotic drug clozapine, and the atypical neuroleptic olanzapine, as well as the sedative drug diphenhydramine and the anxiolytic lorazepam. Diphenhydramine 284-299 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 12709776-2 2003 OBJECTIVE: For this study, the pattern of expression of the protein Fos, a marker of cellular activation, was compared after administration of the typical neuroleptic haloperidol, the antipsychotic drug clozapine, and the atypical neuroleptic olanzapine, as well as the sedative drug diphenhydramine and the anxiolytic lorazepam. Lorazepam 319-328 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 12727327-3 2003 We have addressed this issue by mapping Fos expression in rats acutely injected with diethyldithiocarbamate (DDTC) and correlating these data to neural damage in the hippocampus as determined by pyknotic nuclei count. Ditiocarb 85-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 12727327-3 2003 We have addressed this issue by mapping Fos expression in rats acutely injected with diethyldithiocarbamate (DDTC) and correlating these data to neural damage in the hippocampus as determined by pyknotic nuclei count. Ditiocarb 109-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 12727327-4 2003 In comparison to saline injected rats, DDTC treatment induced a marked Fos expression in most brain regions at 1 and 3 h. In the hippocampus, a high Fos expression was followed by a variable number of pyknotic nuclei at 6 h, depending on the subregion. Ditiocarb 39-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 12727327-4 2003 In comparison to saline injected rats, DDTC treatment induced a marked Fos expression in most brain regions at 1 and 3 h. In the hippocampus, a high Fos expression was followed by a variable number of pyknotic nuclei at 6 h, depending on the subregion. Ditiocarb 39-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 149-152 12727327-5 2003 The data suggest that, in this model of neurotoxicity, c-fos induction does not reflect a cell commitment to die or survive, but rather a cell response to the DDTC-induced oxidative disorder. Ditiocarb 159-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 12717145-9 2003 Midazolam inhibited N(2)O-induced c-Fos expression (a marker of neuronal activation) in the pontine A7 and spinal cord. Midazolam 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 12717145-9 2003 Midazolam inhibited N(2)O-induced c-Fos expression (a marker of neuronal activation) in the pontine A7 and spinal cord. Nitrous Oxide 20-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 12853329-0 2003 Cocaine induces a dose-dependent alteration in the expression of immediate early genes c-fos and SP-1 and in nuclear factor NF-kappabeta in PC12 cells. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 12692480-0 2003 Daily methylphenidate administration attenuates c-fos expression in the striatum of prepubertal rats. Methylphenidate 6-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 12716432-8 2003 The c-fos induction kinetically corresponded to the Elk-1 phosphorylation and was attenuated by antisense oligonucleotides that selectively knocked down Elk-1 proteins. Oligonucleotides 106-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 12686866-10 2003 Actinomycin D suppressed an increase in c-fos mRNA expression 1 hour after middle cerebral artery occlusion as well as in zif268 3 hours after occlusion. Dactinomycin 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 12655462-0 2003 Brain muscarinic receptor subtypes mediating water intake and Fos following cerebroventricular administration of bethanecol in rats. Bethanechol 113-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 12655462-2 2003 OBJECTIVES: To examine whether drinking and brain Fos-immunoreactivity (ir) induced in rats by central administration of bethanecol is reversed by either the preferential M1 antagonist pirenzepine, the M3 antagonist 4-DAMP, or their combination. Bethanechol 121-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 12655462-9 2003 Fos-ir was induced by bethanecol in many brain regions previously implicated in body fluid regulation, including subfornical organ and the magnocellular supraoptic and paraventricular hypothalamic nuclei. Bethanechol 22-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 12655462-10 2003 Induced Fos-ir was substantially but not completely prevented by co-injection of either pirenzepine or 4-DAMP, but their combination did not seem markedly more effective than either alone. Pirenzepine 88-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-11 12655462-10 2003 Induced Fos-ir was substantially but not completely prevented by co-injection of either pirenzepine or 4-DAMP, but their combination did not seem markedly more effective than either alone. 4-diphenylacetoxy-1,1-dimethylpiperidinium 103-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-11 12655462-13 2003 Fos-ir induced in fluid-related brain regions by bethanecol either uses additional receptor type(s) or is less easily blocked than drinking behavior. Bethanechol 49-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 12692480-3 2003 We compared the effects of a single injection of methylphenidate with that of long-term methylphenidate injections (one/day; 14 days) on immediate-early gene expression (c-fos) in the striatum of prepubertal male rats. Methylphenidate 49-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-175 12692480-3 2003 We compared the effects of a single injection of methylphenidate with that of long-term methylphenidate injections (one/day; 14 days) on immediate-early gene expression (c-fos) in the striatum of prepubertal male rats. Methylphenidate 88-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-175 12692480-6 2003 A single injection of methylphenidate (2 or 10 mg/kg) on day 14, following saline treatment for 13 days, caused a dramatic elevation in c-fos expression. Methylphenidate 22-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 12692480-6 2003 A single injection of methylphenidate (2 or 10 mg/kg) on day 14, following saline treatment for 13 days, caused a dramatic elevation in c-fos expression. Sodium Chloride 75-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 12668251-0 2003 Spinal adenosine agonist reduces c-fos and astrocyte activation in dorsal horn of rats with adjuvant-induced arthritis. Adenosine 7-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 12668251-4 2003 In saline-treated AIA animals the increase in c-Fos expression seen in laminae I-IV was accompanied by an increase in glial fibrillary acidic protein immunoreactivity, indicative of astrocyte activation. Sodium Chloride 3-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 12670703-7 2003 The complexes formed with labeled AP1/E box oligonucleotide were reduced or supershifted with antisera to Fos family, c-Fos, Fra-2, and Jun D. Oligonucleotides 44-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 12670703-7 2003 The complexes formed with labeled AP1/E box oligonucleotide were reduced or supershifted with antisera to Fos family, c-Fos, Fra-2, and Jun D. Oligonucleotides 44-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 12671717-11 2003 The two COX inhibitors did not change c-myc expression, significantly decreased the expression of Egr-1, and differentially altered expression of c-fos; aspirin did not change, but celecoxib dramatically decreased the levels of c-fos-mRNA. Celecoxib 181-190 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 228-233 12676367-0 2003 Rewarding injections of the cholinergic agonist carbachol into the ventral tegmental area induce locomotion and c-Fos expression in the retrosplenial area and supramammillary nucleus. Carbachol 48-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 12676367-2 2003 To determine what brain regions are activated by such rewarding injections we studied the expression of the transcription factor c-Fos in local and distant brain regions following ventral tegmental injections of carbachol in rats. Carbachol 212-221 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 12676367-5 2003 Ventral tegmental injections of carbachol induced c-Fos expression throughout the brain. Carbachol 32-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 12505866-9 2003 Levels of Fos-ir were highest in fluid-depleted rats that drank water and sodium. Water 64-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-13 12505866-9 2003 Levels of Fos-ir were highest in fluid-depleted rats that drank water and sodium. Sodium 74-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-13 12721110-0 2003 Effects of U-50488H and U-50488H withdrawal on c-fos expression in the rat paraventricular nucleus. 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 12721110-3 2003 In the present work, we have studied the expression of Fos during acute and chronic administration of the kappa-opioid receptor agonist U-50488H and after U-5088H withdrawal in the rat hypothalamic paraventricular nucleus (PVN). 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer 136-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 12721110-15 2003 Using immunohistochemical staining of Fos, present results indicate that acute administration of U-50488H produced an increase in Fos expression in the PVN and in the noradrenergic A(1) and A(2) cell groups. 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer 97-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 12721110-15 2003 Using immunohistochemical staining of Fos, present results indicate that acute administration of U-50488H produced an increase in Fos expression in the PVN and in the noradrenergic A(1) and A(2) cell groups. 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer 97-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-133 12721110-16 2003 Moreover, when double-label immunohistochemistry was used to identify Fos and catecholaminergic-positive neurons in the brainstem, it was found that catecholaminergic-positive neurons in the NTS and VLM showed a significant increase in Fos expression in response to acute U-50488H injection. 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer 272-280 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 12721110-16 2003 Moreover, when double-label immunohistochemistry was used to identify Fos and catecholaminergic-positive neurons in the brainstem, it was found that catecholaminergic-positive neurons in the NTS and VLM showed a significant increase in Fos expression in response to acute U-50488H injection. 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer 272-280 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 236-239 12721110-17 2003 Chronic application of U-50488H leads to the development of tolerance towards their effects on Fos expression in the PVN as well as in the NTS and VLM. 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer 23-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 12752381-5 2003 This perfusion with muscimol at 50 micro m produced FOS-like immunoreactivity in 37% of the orexin neurons located in the tuberal part of the hypothalamus, whereas the FOS-like immunoreactivity was sparse in orexin neurons of the sleeping control rats (P = 0.001 by Mann-Whitney U-test). Muscimol 20-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 12663692-4 2003 The binding activity of GRE activating protein-1 (AP-1) site and cross-competition analysis demonstrated specific pattern of expression during the ageing and DEX treatment, suggesting that GR modulates the activity of transcription factors AP-1 (Fos/Jun proteins) through protein-protein interaction. Dexamethasone 158-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 246-249 12453873-3 2003 Electrophoretic mobility shift assays identified Nrf2, MafG, ATF3, and Jun and Fos family members as StRE-binding proteins; binding of Nrf2, MafG, and ATF3 was increased in response to heme. Heme 185-189 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 14761536-10 2003 (3) There were much more deep-colored c-fos positive cells in CA3 area of hippocampus in low dose of Pb-exposed with Gastrodia elata + E-gelatin group. Lead 101-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 14967940-3 2003 In the present study, 2DG-induced Fos expression was examined in the solitary tract nucleus (NTS), hypothalamic paraventricular nucleus (PVN), raphe obscurus nucleus (ROb) and raphe pallidus nucleus (RPa), which have been previously suggested to be involved in glucoprivation-induced suppression of LH secretion in female rats. Deoxyglucose 22-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 14967940-9 2003 Some Fos-li cells (8.3%) had tyrosine hydroxylase-like immunoreactivities in the NTS in 2DG-treated OVX+E(2) rats. Deoxyglucose 88-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 5-8 12650967-0 2003 Nitrous oxide-induced c-Fos expression in the rat brain. Nitrous Oxide 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 12650967-1 2003 Induction of c-Fos has previously been used to map locations of cells in the central nervous system (CNS) that are activated by ethanol administration. Ethanol 128-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 12650967-5 2003 N(2)O administration produced significant (P<0.05) dose-related increases of c-Fos expression in several forebrain regions, including the hypothalamic supraoptic and paraventricular nuclei, the thalamic paraventricular nucleus, the amygdala, and in retrosplenial cortex. Nitrous Oxide 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 12650967-6 2003 In the midbrain, N(2)O caused significant dose-related c-Fos expression in the Edinger-Westphal nucleus. Nitrous Oxide 17-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 12650967-7 2003 Finally, the pontine locus coeruleus, and two medullary regions, the nucleus of the solitary tract and ventrolateral medulla, also showed significant dose-related N(2)O-induced c-Fos expression. Nitrous Oxide 163-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 177-182 12650967-9 2003 The overlapping pattern of c-Fos induced by ethanol and N(2)O suggests that these drugs may cause comparable central activity by acting on similar neuronal pathways. Ethanol 44-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 12650967-9 2003 The overlapping pattern of c-Fos induced by ethanol and N(2)O suggests that these drugs may cause comparable central activity by acting on similar neuronal pathways. Nitrous Oxide 56-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 12650968-2 2003 In conscious female Wistar rats, c-fos expression was induced by continuous intravesical infusion of saline or 0.1% acetic acid. Sodium Chloride 101-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 12650968-2 2003 In conscious female Wistar rats, c-fos expression was induced by continuous intravesical infusion of saline or 0.1% acetic acid. Acetic Acid 116-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 12650968-5 2003 Compared with saline infusion, acetic acid infusion provoked irritative bladder responses characterized by a marked increase in the frequency of bladder contractions, and induced a significant increase in the number of Fos-positive cells in both L1 and L6 of the spinal cord. Acetic Acid 31-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 219-222 12650968-6 2003 Following acetic acid infusion, there was a significant increase in the number of Fos-positive cells in all coronal sections of the PAG compared with saline infusion, especially in the caudal part of the PAG. Acetic Acid 10-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 12618353-7 2003 injection of carrageenan, the number of Fos-like immunoreactive (Fos-LI) neurons was significantly increased in all layers of ipsilateral spinal cord at L(4)-L(5) with the higher density in laminae I-II and V-VI. Carrageenan 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 12618353-7 2003 injection of carrageenan, the number of Fos-like immunoreactive (Fos-LI) neurons was significantly increased in all layers of ipsilateral spinal cord at L(4)-L(5) with the higher density in laminae I-II and V-VI. Carrageenan 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 12618353-8 2003 Intrathecally pre-administered KYNA (10 nmol) significantly reduced the total number of carrageenan-induced Fos-LI neurons with more apparent reduction in laminae I-II and IV-V. Pre-coapplication of 10 nmol KYNA and EA of bilateral "Zu-San-Li" and "Kun-Lun" acupoints, the numbers of carrageenan-induced Fos-LI neurons in laminae I-II and V-VI further reduced. Kynurenic Acid 31-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 12618353-8 2003 Intrathecally pre-administered KYNA (10 nmol) significantly reduced the total number of carrageenan-induced Fos-LI neurons with more apparent reduction in laminae I-II and IV-V. Pre-coapplication of 10 nmol KYNA and EA of bilateral "Zu-San-Li" and "Kun-Lun" acupoints, the numbers of carrageenan-induced Fos-LI neurons in laminae I-II and V-VI further reduced. Kynurenic Acid 31-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 304-307 12618353-8 2003 Intrathecally pre-administered KYNA (10 nmol) significantly reduced the total number of carrageenan-induced Fos-LI neurons with more apparent reduction in laminae I-II and IV-V. Pre-coapplication of 10 nmol KYNA and EA of bilateral "Zu-San-Li" and "Kun-Lun" acupoints, the numbers of carrageenan-induced Fos-LI neurons in laminae I-II and V-VI further reduced. Carrageenan 88-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 12618353-8 2003 Intrathecally pre-administered KYNA (10 nmol) significantly reduced the total number of carrageenan-induced Fos-LI neurons with more apparent reduction in laminae I-II and IV-V. Pre-coapplication of 10 nmol KYNA and EA of bilateral "Zu-San-Li" and "Kun-Lun" acupoints, the numbers of carrageenan-induced Fos-LI neurons in laminae I-II and V-VI further reduced. Carrageenan 88-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 304-307 12618353-9 2003 The level of Fos expression in the spinal cord induced by carrageenan was significantly lower compared with that of i.t. Carrageenan 58-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 12646303-0 2003 Effect of indomethacin on the c-fos expression in AVP and TH neurons in rat brain induced by lipopolysaccharide. Indomethacin 10-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 12646303-1 2003 The aim of the present study was to investigate the effect of indomethacin on the Fos expression in arginine vasopressin (AVP)-containing neurons in the hypothalamus and tyrosine hydroxylase (TH)-containing neurons in the locus coeruleus (LC) using dual-labeled immunohistochemistry. Indomethacin 62-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 12646303-7 2003 Indomethacin prevented the Fos expression induced only by a low dose of LPS, but not by a high dose of LPS. Indomethacin 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 12618300-0 2003 Asymmetric Fos labeling in lobule X of the cerebellum following transtympanic tetrodotoxin (TTX) in the rat. transtympanic tetrodotoxin 64-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 12618300-0 2003 Asymmetric Fos labeling in lobule X of the cerebellum following transtympanic tetrodotoxin (TTX) in the rat. Tetrodotoxin 92-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 12618300-3 2003 The granular and molecular layers as well as the medial cerebellar nucleus ipsilateral to TTX treatment contained elevated levels of Fos relative to the same regions contralaterally and when compared to controls receiving equal volume injections of vehicle. Tetrodotoxin 90-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-136 12648194-0 2003 Inhibitory effect of propofol on ketamine-induced c-Fos expression in the rat posterior cingulate and retrosplenial cortices is mediated by GABAA receptor activation. Propofol 21-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 12648194-0 2003 Inhibitory effect of propofol on ketamine-induced c-Fos expression in the rat posterior cingulate and retrosplenial cortices is mediated by GABAA receptor activation. Ketamine 33-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 12648194-2 2003 We previously demonstrated that ketamine induced marked c-Fos (c-fos protein) expression in the rat PC/RS, which was inhibited by propofol, and the expression was closely related to ketamine-induced abnormal behavior. Ketamine 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 12648194-2 2003 We previously demonstrated that ketamine induced marked c-Fos (c-fos protein) expression in the rat PC/RS, which was inhibited by propofol, and the expression was closely related to ketamine-induced abnormal behavior. Ketamine 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 12648194-2 2003 We previously demonstrated that ketamine induced marked c-Fos (c-fos protein) expression in the rat PC/RS, which was inhibited by propofol, and the expression was closely related to ketamine-induced abnormal behavior. Propofol 130-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 12648194-2 2003 We previously demonstrated that ketamine induced marked c-Fos (c-fos protein) expression in the rat PC/RS, which was inhibited by propofol, and the expression was closely related to ketamine-induced abnormal behavior. Propofol 130-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 12648194-2 2003 We previously demonstrated that ketamine induced marked c-Fos (c-fos protein) expression in the rat PC/RS, which was inhibited by propofol, and the expression was closely related to ketamine-induced abnormal behavior. Ketamine 182-190 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 12648194-2 2003 We previously demonstrated that ketamine induced marked c-Fos (c-fos protein) expression in the rat PC/RS, which was inhibited by propofol, and the expression was closely related to ketamine-induced abnormal behavior. Ketamine 182-190 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 12648194-7 2003 RESULTS: Ketamine induced marked c-Fos expression in the PC/RS (171 +/- 9/0.4 mm2), which was significantly inhibited by propofol (5 +/- 5/0.4 mm2). Ketamine 9-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 12648194-7 2003 RESULTS: Ketamine induced marked c-Fos expression in the PC/RS (171 +/- 9/0.4 mm2), which was significantly inhibited by propofol (5 +/- 5/0.4 mm2). Propofol 121-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 12648194-9 2003 CONCLUSION: These results demonstrate that the inhibitory effect of propofol on ketamine-induced c-Fos expression in the PC/RS is mediated by GABAA receptor activation, and suggests that ketamine-induced psychoneuronal adverse effects may be suppressed by propofol via the activation of GABAA receptors. Propofol 68-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 12648194-9 2003 CONCLUSION: These results demonstrate that the inhibitory effect of propofol on ketamine-induced c-Fos expression in the PC/RS is mediated by GABAA receptor activation, and suggests that ketamine-induced psychoneuronal adverse effects may be suppressed by propofol via the activation of GABAA receptors. Ketamine 80-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 12648194-9 2003 CONCLUSION: These results demonstrate that the inhibitory effect of propofol on ketamine-induced c-Fos expression in the PC/RS is mediated by GABAA receptor activation, and suggests that ketamine-induced psychoneuronal adverse effects may be suppressed by propofol via the activation of GABAA receptors. Ketamine 187-195 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 12648194-9 2003 CONCLUSION: These results demonstrate that the inhibitory effect of propofol on ketamine-induced c-Fos expression in the PC/RS is mediated by GABAA receptor activation, and suggests that ketamine-induced psychoneuronal adverse effects may be suppressed by propofol via the activation of GABAA receptors. Propofol 256-264 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 12632255-5 2003 Neither insulin nor dexamethasone alone induced c- fos mRNA but stimulation of c- fos expression by EGF plus estradiol occurred earlier in the presence of insulin, and was augmented by preincubation of cells with dexamethasone. Dexamethasone 213-226 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-85 12650969-7 2003 The number of Fos-LI neurons observed when the MEK inhibitor, U0126, was administered was significantly suppressed compared to the DMSO- (vehicle control) administered group. U 0126 62-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 12650969-7 2003 The number of Fos-LI neurons observed when the MEK inhibitor, U0126, was administered was significantly suppressed compared to the DMSO- (vehicle control) administered group. Dimethyl Sulfoxide 131-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 12650971-1 2003 A spinal c-Fos protein study in the rat under carrageenin inflammation. Carrageenan 46-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 12650971-2 2003 We have previously shown that RB101, a dual inhibitor of enkephalin-degrading enzymes, decreased carrageenin-evoked c-Fos protein expression at the spinal cord level in awake rats. RB 101 30-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 12650971-2 2003 We have previously shown that RB101, a dual inhibitor of enkephalin-degrading enzymes, decreased carrageenin-evoked c-Fos protein expression at the spinal cord level in awake rats. Carrageenan 97-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 12650971-6 2003 c-Fos protein-immunoreactivity (c-Fos-IR) was evaluated as the number of c-Fos-IR nuclei in the lumbar spinal cord 90 min after carrageenin. Carrageenan 128-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 12650971-6 2003 c-Fos protein-immunoreactivity (c-Fos-IR) was evaluated as the number of c-Fos-IR nuclei in the lumbar spinal cord 90 min after carrageenin. Carrageenan 128-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 12650971-6 2003 c-Fos protein-immunoreactivity (c-Fos-IR) was evaluated as the number of c-Fos-IR nuclei in the lumbar spinal cord 90 min after carrageenin. Carrageenan 128-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 12632255-6 2003 EGF + E(2)-induced c- fos mRNA was completely abolished by actinomycin D, suggesting that transcription is the major mechanism for c- fos induction by E(2) + EGF. Dactinomycin 59-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-25 12632255-6 2003 EGF + E(2)-induced c- fos mRNA was completely abolished by actinomycin D, suggesting that transcription is the major mechanism for c- fos induction by E(2) + EGF. Dactinomycin 59-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-137 12576146-2 2003 Induction of c-fos mRNA due to 4-AP-elicited seizures was ascertained by reverse transcription polymerase chain reaction in samples of the neocortex. 4-Aminopyridine 31-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 12650971-9 2003 significantly reduced the total number of carrageenin-evoked c-Fos-IR nuclei (30% reduction, P<0.01). Carrageenan 42-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 12531432-4 2003 In addition, Ti-induced SRE activation was shown to be dramatically repressed by RacN17, a dominant negative mutant of Rac1, suggesting that Rac GTPase is essential for the signalling of Ti to c-fos SRE. Titanium 13-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-198 12653981-9 2003 However, running-wheel training under the influence of cocaine enhanced the c-fos response to a subsequent cocaine challenge selectively in parts of the caudal sensorimotor striatum. Cocaine 55-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 12653981-9 2003 However, running-wheel training under the influence of cocaine enhanced the c-fos response to a subsequent cocaine challenge selectively in parts of the caudal sensorimotor striatum. Cocaine 107-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 12679565-6 2003 In addition, BV pretreatment into the Zusanli acupoint markedly inhibited spinal cord Fos expression induced by formalin injection. Formaldehyde 112-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 12566172-6 2003 The aim of the present study was to investigate the effects of serotonin depletion on acute CRF-induced c-fos expression, corticosterone levels and behavioural responses. Serotonin 63-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 12576146-7 2003 Treatment with either antagonist did not induce by itself c-fos expression, with the exception of amantadine, which caused slight Fos induction in the neocortex. Amantadine 98-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-133 12576146-10 2003 MK-801, ketamine and dextrometorphan decreased significantly Fos immunoreactivity also in area CA3. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 12586446-4 2003 All drugs but one (mCPP) also increased Fos expression in the basolateral and medial amygdala, the dorsomedial hypothalamus, cingulate cortex, and parts of the motor cortex. 1-(3-chlorophenyl)piperazine 19-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 12581847-10 2003 Cocaine-induced alterations in expression of c-fos and preprodynorphin mRNAs measured by TaqMan were confirmed by ribonuclease protection assay. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 12576146-10 2003 MK-801, ketamine and dextrometorphan decreased significantly Fos immunoreactivity also in area CA3. Ketamine 8-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 12576146-10 2003 MK-801, ketamine and dextrometorphan decreased significantly Fos immunoreactivity also in area CA3. Dextromethorphan 21-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 12588467-1 2003 Challenge of the rat gastric mucosa with 0.5 mol L(-1) HCl activates nitrergic neurons in the myenteric plexus as visualized by c-Fos immunohistochemistry. Hydrochloric Acid 55-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-133 12552203-4 2003 RESULTS: Dexmedetomidine induced a qualitatively similar pattern of c-Fos expression in rats as seen during normal NREM sleep, a decrease in the locus ceruleus (LC) and tuberomammillary nucleus (TMN) and an increase in the ventrolateral preoptic nucleus (VLPO). Dexmedetomidine 9-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 12552203-7 2003 VLPO lesions and gabazine pretreatment altered c-Fos expression in the TMN but in not the LC after dexmedetomidine administration, indicating a hierarchical sequence of changes. gabazine 17-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 12598416-0 2003 Activation of c-fos expression in the heart after morphine but not U-50,488H withdrawal. Morphine 50-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 12598416-2 2003 In the present work we have studied in the heart the expression of Fos, the protein product of the c-fos proto-oncogene and the adaptive changes in noradrenergic neurons after naloxone or nor-binaltorphimine (nor-BNI) administration to morphine or U-50,488H pretreated rats. norbinaltorphimine 209-216 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 12598416-2 2003 In the present work we have studied in the heart the expression of Fos, the protein product of the c-fos proto-oncogene and the adaptive changes in noradrenergic neurons after naloxone or nor-binaltorphimine (nor-BNI) administration to morphine or U-50,488H pretreated rats. Morphine 236-244 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 12598416-2 2003 In the present work we have studied in the heart the expression of Fos, the protein product of the c-fos proto-oncogene and the adaptive changes in noradrenergic neurons after naloxone or nor-binaltorphimine (nor-BNI) administration to morphine or U-50,488H pretreated rats. u-50 248-252 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 12598416-2 2003 In the present work we have studied in the heart the expression of Fos, the protein product of the c-fos proto-oncogene and the adaptive changes in noradrenergic neurons after naloxone or nor-binaltorphimine (nor-BNI) administration to morphine or U-50,488H pretreated rats. 488h 253-257 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 12598416-12 2003 Using immunohistochemical staining of Fos, present results indicate that morphine withdrawal induced marked Fos immunoreactivity (Fos-IR) within the cardiomyocyte nuclei. Morphine 73-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 12598416-12 2003 Using immunohistochemical staining of Fos, present results indicate that morphine withdrawal induced marked Fos immunoreactivity (Fos-IR) within the cardiomyocyte nuclei. Morphine 73-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 12598416-12 2003 Using immunohistochemical staining of Fos, present results indicate that morphine withdrawal induced marked Fos immunoreactivity (Fos-IR) within the cardiomyocyte nuclei. Morphine 73-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 12598416-13 2003 Moreover, Western blots analysis revealed a peak expression of c-fos in right and left ventricle after naloxone induced withdrawal in parallel with an increase in noradrenaline (NA) turnover. Naloxone 103-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 12598416-18 2003 These results demonstrated that morphine withdrawal induces the expression of Fos protein, as well as an enhancement of noradrenergic activity in the heart. Morphine 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 12603266-5 2003 Animals treated with SKF 38393 showed dose-dependent, massive Fos increases in the motor, somatosensory, auditory, visual and limbic regions of the cerebral cortex, ipsilaterally to the injected striatum. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 21-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 12583883-0 2003 Differential changes in Fos-immunoreactivity at the auditory brainstem after chronic injections of salicylate in rats. Salicylates 99-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 12583883-2 2003 In this study, we revealed the distribution of active cells in the rat auditory brainstem by staining an activity marker Fos-protein after multiple daily injections of salicylate. Salicylates 168-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-124 12589382-0 2003 Enhanced morphine preference following prolonged abstinence: association with increased Fos expression in the extended amygdala. Morphine 9-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 12589382-3 2003 To determine brain regions involved in this behavior, we examined neural activation (as indexed by Fos-like proteins) induced by a morphine-conditioned place preference test. Morphine 131-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 12589382-4 2003 Placebo-pretreated (P) morphine-conditioned rats showed significantly elevated Fos in the anterior cingulate cortex (Cg), nucleus accumbens core (Ac-C) and shell (Ac-S), ventral lateral and dorsal lateral bed nucleus of the stria terminialis (BNST-VL and -DL), and central and basolateral amygdala nuclei (ACE, ABL) when compared to nonconditioned P rats. Morphine 23-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 12589382-5 2003 Chronically morphine-pretreated (M) rats that exhibited enhanced morphine preference 5 weeks after morphine withdrawal showed significantly greater Fos in all the same areas except the BNST-DL relative to conditioned P or nonconditioned M rats. Morphine 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-151 12623225-0 2003 Regional differences in naloxone modulation of Delta(9)-THC induced Fos expression in rat brain. Naloxone 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 12623225-0 2003 Regional differences in naloxone modulation of Delta(9)-THC induced Fos expression in rat brain. Dronabinol 56-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 12623225-7 2003 Results showed that naloxone inhibited THC-induced Fos immunoreactivity in several key brain regions including the ventral tegmental area, ventromedial and dorsomedial hypothalamus, central caudate-putamen and ventrolateral periaqueductal grey. Naloxone 20-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 12623225-7 2003 Results showed that naloxone inhibited THC-induced Fos immunoreactivity in several key brain regions including the ventral tegmental area, ventromedial and dorsomedial hypothalamus, central caudate-putamen and ventrolateral periaqueductal grey. Dronabinol 39-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 12623225-8 2003 Conversely, naloxone and THC had an additive effect on Fos immunoreactivity in the central nucleus of the amygdala, the bed nucleus of the stria terminalis (lateral division), the insular cortex, and the paraventricular nucleus of the thalamus. Naloxone 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 12623225-8 2003 Conversely, naloxone and THC had an additive effect on Fos immunoreactivity in the central nucleus of the amygdala, the bed nucleus of the stria terminalis (lateral division), the insular cortex, and the paraventricular nucleus of the thalamus. Dronabinol 25-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 12623225-10 2003 The inhibitory effects of naloxone on THC-induced ventral tegmentum, hypothalamic and periaqueductal grey Fos expression point to these structures as key sites involved in cannabinoid-opioid interactions. Naloxone 26-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 12623225-10 2003 The inhibitory effects of naloxone on THC-induced ventral tegmentum, hypothalamic and periaqueductal grey Fos expression point to these structures as key sites involved in cannabinoid-opioid interactions. Dronabinol 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 12623225-10 2003 The inhibitory effects of naloxone on THC-induced ventral tegmentum, hypothalamic and periaqueductal grey Fos expression point to these structures as key sites involved in cannabinoid-opioid interactions. Cannabinoids 172-183 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 12538785-6 2003 In addition, the hypoxic respiratory depression is clearly emphasized after in utero exposure to caffeine and coincides with an increased Fos expression in the area postrema and nucleus raphe obscurus, two structures in which it is not increased in the absence of caffeine. Caffeine 264-272 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-141 12543232-1 2003 The effects of intrathecal nociceptin (NOCI) on the nociceptive behavior (biting, scratching and licking; BSL) and the spinal Fos expression induced by intrathecal administration of N-methyl-D-aspartate (NMDA, 4 microg/rat) or alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate (AMPA, 2 microg/rat) were studied. N-Methylaspartate 182-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-129 12588467-6 2003 Pre-treatment with capsaicin or hexamethonium or a combination of both pre-treatments reduced HCl-induced c-Fos expression by 54, 66 and 63%, respectively. Capsaicin 19-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 12588467-6 2003 Pre-treatment with capsaicin or hexamethonium or a combination of both pre-treatments reduced HCl-induced c-Fos expression by 54, 66 and 63%, respectively. Hexamethonium 32-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 12588467-6 2003 Pre-treatment with capsaicin or hexamethonium or a combination of both pre-treatments reduced HCl-induced c-Fos expression by 54, 66 and 63%, respectively. Hydrochloric Acid 94-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 12573543-0 2003 Prolonged expression of c-Fos and c-Jun in the cerebral cortex of rats after deltamethrin treatment. decamethrin 77-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 12573543-1 2003 In this study we investigated the effects of deltamethrin on the expression of c-Fos and c-Jun in the cerebral cortex of rats. decamethrin 45-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 12573543-2 2003 Immunohistochemical analysis demonstrated that the immunoreactivity for c-Fos was markedly increased in the cerebral cortex 5 h after deltamethrin treatment, and maintained at an increased level at 24 h, even though little immunoreactivity for c-Fos was seen in the same brain region of control rats. decamethrin 134-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 12573543-2 2003 Immunohistochemical analysis demonstrated that the immunoreactivity for c-Fos was markedly increased in the cerebral cortex 5 h after deltamethrin treatment, and maintained at an increased level at 24 h, even though little immunoreactivity for c-Fos was seen in the same brain region of control rats. decamethrin 134-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 244-249 12576128-8 2003 DFEN induced Fos-ir in brain regions analogous to those reported in rats and, also, as in rats, DFEN pretreatments reduced substantially the Fos-ir induced by acute DFEN in regions including central nucleus of amygdala, bed nucleus of stria terminalis, and the paraventricular hypothalamus (magno- and parvocellular divisions; PVN). Dexfenfluramine 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 12576128-8 2003 DFEN induced Fos-ir in brain regions analogous to those reported in rats and, also, as in rats, DFEN pretreatments reduced substantially the Fos-ir induced by acute DFEN in regions including central nucleus of amygdala, bed nucleus of stria terminalis, and the paraventricular hypothalamus (magno- and parvocellular divisions; PVN). Dexfenfluramine 96-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 12576128-8 2003 DFEN induced Fos-ir in brain regions analogous to those reported in rats and, also, as in rats, DFEN pretreatments reduced substantially the Fos-ir induced by acute DFEN in regions including central nucleus of amygdala, bed nucleus of stria terminalis, and the paraventricular hypothalamus (magno- and parvocellular divisions; PVN). Dexfenfluramine 96-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 12576128-8 2003 DFEN induced Fos-ir in brain regions analogous to those reported in rats and, also, as in rats, DFEN pretreatments reduced substantially the Fos-ir induced by acute DFEN in regions including central nucleus of amygdala, bed nucleus of stria terminalis, and the paraventricular hypothalamus (magno- and parvocellular divisions; PVN). Dexfenfluramine 96-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 12576128-8 2003 DFEN induced Fos-ir in brain regions analogous to those reported in rats and, also, as in rats, DFEN pretreatments reduced substantially the Fos-ir induced by acute DFEN in regions including central nucleus of amygdala, bed nucleus of stria terminalis, and the paraventricular hypothalamus (magno- and parvocellular divisions; PVN). Dexfenfluramine 96-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 12505681-1 2003 In this study we examined the effects of the glutamate metabotropic subtype 5 (mGlu5) receptor antagonist 2-methyl-6-(phenylethynyl)-pyridine (MPEP) on Fos expression in the spinal cord in a model of visceral pain in the rat. 6-methyl-2-(phenylethynyl)pyridine 106-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-155 12505681-1 2003 In this study we examined the effects of the glutamate metabotropic subtype 5 (mGlu5) receptor antagonist 2-methyl-6-(phenylethynyl)-pyridine (MPEP) on Fos expression in the spinal cord in a model of visceral pain in the rat. 6-methyl-2-(phenylethynyl)pyridine 143-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-155 12505681-2 2003 We show that noxious stimulation increases the number of Fos-positive neurons in the dorsal horn of the thoracic and lumbar spinal cord, and that pretreatment with MPEP significantly reduces the number of Fos-positive neurons in these areas. 6-methyl-2-(phenylethynyl)pyridine 164-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 12505681-2 2003 We show that noxious stimulation increases the number of Fos-positive neurons in the dorsal horn of the thoracic and lumbar spinal cord, and that pretreatment with MPEP significantly reduces the number of Fos-positive neurons in these areas. 6-methyl-2-(phenylethynyl)pyridine 164-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 205-208 14562759-7 2003 At this stage, the oxygen concentration in cerebral tissue fell to lower than 10(-7) M. These data suggest that the decline in oxidative phosphorylation might be a trigger for the induction of c-fos mRNA. Oxygen 19-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-198 12520169-0 2003 Effect of Puerariae radix on c-Fos expression in hippocampus of alcohol-intoxicated juvenile rats. Alcohols 64-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 12520169-3 2003 In the present study, the effect of the aqueous extract of Puerariae radix on the expression of c-Fos, an immediate early gene whose expression is sometimes used as a marker for stimulus-induced changes in the metabolic activity of neurons, in the hippocampus of acutely alcohol-intoxicated juvenile rats was investigated via immunohistochemistry. Alcohols 271-278 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 12520169-6 2003 From the results, it was demonstrated that alcohol administration significantly decreases the number of Fos-positive cells in the various regions of the hippocampus, and Puerariae radix treatment inhibits the alcohol-induced suppression of the expression of Fos in the hippocampus in a dose-dependent manner. Alcohols 43-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 12520169-6 2003 From the results, it was demonstrated that alcohol administration significantly decreases the number of Fos-positive cells in the various regions of the hippocampus, and Puerariae radix treatment inhibits the alcohol-induced suppression of the expression of Fos in the hippocampus in a dose-dependent manner. Alcohols 43-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 258-261 12520169-6 2003 From the results, it was demonstrated that alcohol administration significantly decreases the number of Fos-positive cells in the various regions of the hippocampus, and Puerariae radix treatment inhibits the alcohol-induced suppression of the expression of Fos in the hippocampus in a dose-dependent manner. Alcohols 209-216 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 12520169-6 2003 From the results, it was demonstrated that alcohol administration significantly decreases the number of Fos-positive cells in the various regions of the hippocampus, and Puerariae radix treatment inhibits the alcohol-induced suppression of the expression of Fos in the hippocampus in a dose-dependent manner. Alcohols 209-216 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 258-261 12534973-8 2003 Morphine withdrawal induced the expression of Fos in the PVN and NTS/VLM, which indicates an activation of neurons in these nuclei. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 12725847-0 2003 Antinociceptive effects of RB101(S), a complete inhibitor of enkephalin-catabolizing enzymes, are enhanced by (+)-HA966, a functional NMDA receptor antagonist: a c-Fos study in the rat spinal cord. RB 101 27-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 12725847-2 2003 One hour 30min after intraplantar carrageenan in awake rats, c-Fos immunoreactive (c-Fos-IR) nuclei were preferentially located in the laminae I-II and V-VI of the spinal dorsal horn, i.e., spinal areas containing numerous neurons responding exclusively, or not, to peripheral nociceptive stimuli. Carrageenan 34-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 12725847-5 2003 Two highest doses of RB101(S) (20 and 40mg/kg) significantly reduced the number of carrageenan-evoked c-Fos-IR nuclei in both superficial I-II (32+/-7% and 36+/-5% reduction, respectively, P<0.05 for both) and deep V-VI (42+/-6% and 61+/-2% reduction, respectively, P<0.001 for both) laminae. Carrageenan 83-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 12573543-5 2003 Since the persistent expression of c-Fos and c-Jun is unusual, and has been reported before in conditions involving neurodegeneration, our results are consistent with a model that deltamethrin induces neurodegeneration through a glutamate-dependent pathway. decamethrin 180-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 12573543-5 2003 Since the persistent expression of c-Fos and c-Jun is unusual, and has been reported before in conditions involving neurodegeneration, our results are consistent with a model that deltamethrin induces neurodegeneration through a glutamate-dependent pathway. Glutamic Acid 229-238 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 12566981-12 2003 The results of our studies show that treatments with 7-nitroindazole, a specific inhibitor of nitric oxide synthase known to attenuate nitric oxide, oxidative DNA lesions, and necrosis, increase intact c-fos mRNA levels after stroke. 7-nitroindazole 53-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 202-207 12520409-3 2003 Following nicotine and capsaicin, there was a significant increase in fos-like immunoreactivity (FLI) compared with controls in the following areas: nucleus of the solitary tract from the level of the pyramidal decussation caudally to the level of the area postrema rostrally; dorsomedial aspect of trigeminal subnucleus caudalis (Vc); and paratrigeminal islands interspersed in the spinal trigeminal tract. Nicotine 10-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 12520409-3 2003 Following nicotine and capsaicin, there was a significant increase in fos-like immunoreactivity (FLI) compared with controls in the following areas: nucleus of the solitary tract from the level of the pyramidal decussation caudally to the level of the area postrema rostrally; dorsomedial aspect of trigeminal subnucleus caudalis (Vc); and paratrigeminal islands interspersed in the spinal trigeminal tract. Capsaicin 23-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 12621163-7 2003 Pitavastatin also inhibited lysoPC-induced c-fos mRNA expression. pitavastatin 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 12566981-12 2003 The results of our studies show that treatments with 7-nitroindazole, a specific inhibitor of nitric oxide synthase known to attenuate nitric oxide, oxidative DNA lesions, and necrosis, increase intact c-fos mRNA levels after stroke. nitric 94-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 202-207 12566981-12 2003 The results of our studies show that treatments with 7-nitroindazole, a specific inhibitor of nitric oxide synthase known to attenuate nitric oxide, oxidative DNA lesions, and necrosis, increase intact c-fos mRNA levels after stroke. Nitric Oxide 94-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 202-207 12508050-17 2003 In normal myotubes, 10 micro M nifedipine, but not ryanodine, inhibited c-jun and c-fos mRNA increase after K(+) depolarization. Nifedipine 31-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 14526427-0 2003 Effect of levodopa chronic administration on behavioral changes and fos expression in basal ganglia in rat model of PD. Levodopa 10-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 14526427-5 2003 Fos positive nuclei in the CPU and GP were increased by levodopa acute administration, and more remarkably in the CPU, but not in the cerebral cortex. Levodopa 56-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 12527992-1 2003 We used the rats in which one olfactory nerve had been transected and observed the odor (Propionic acid) -induced c-Fos immunoreactivity in the bulb at different times (2, 4, 8 weeks) after nerve transection. propionic acid 89-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 12558986-10 2003 The increase in c-fos was kinetically correlated well with the CREB phosphorylation and blocked by MPEP and the CREB antisense oligonucleotides. MPEP 99-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 12558986-10 2003 The increase in c-fos was kinetically correlated well with the CREB phosphorylation and blocked by MPEP and the CREB antisense oligonucleotides. Oligonucleotides 127-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 12544632-8 2003 A significantly higher number of c-fos expressing neurons were observed in the periventricular region of the steroid-2 than the control and vehicle groups, indicating enhanced neuronal activity after nandrolone decanoate treatment. Steroids 109-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 12544632-8 2003 A significantly higher number of c-fos expressing neurons were observed in the periventricular region of the steroid-2 than the control and vehicle groups, indicating enhanced neuronal activity after nandrolone decanoate treatment. Nandrolone Decanoate 200-220 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 12496936-2 2003 Previous work in rats has shown that cues associated with morphine, cocaine, nicotine or palatable food can elicit enhanced expression of the immediate-early gene product Fos in discrete brain regions. Morphine 58-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-174 12496936-2 2003 Previous work in rats has shown that cues associated with morphine, cocaine, nicotine or palatable food can elicit enhanced expression of the immediate-early gene product Fos in discrete brain regions. Cocaine 68-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-174 12496936-2 2003 Previous work in rats has shown that cues associated with morphine, cocaine, nicotine or palatable food can elicit enhanced expression of the immediate-early gene product Fos in discrete brain regions. Nicotine 77-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-174 12617307-1 2003 In situ hybridization using oligonucleotide probes was used to study the effects of intrastriatal microinjection of corticoliberin on the expression of the early genes c-fos, jun B, c-jun, and NGFIA in the rat brain. Oligonucleotides 28-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-173 12890524-0 2003 Environmental context and drug history modulate amphetamine-induced c-fos mRNA expression in the basal ganglia, central extended amygdala, and associated limbic forebrain. Amphetamine 48-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 12890524-2 2003 When given in a novel test environment amphetamine produces greater levels of c-fos and arc mRNA expression in many brain regions relative to when it is given in the home cage. Amphetamine 39-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 12890524-3 2003 The purpose of the current study was to determine if environment and drug history interact to influence amphetamine-induced c-fos mRNA expression. Amphetamine 104-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 12890524-8 2003 In most brain regions amphetamine given in the Novel environment produced greater c-fos mRNA expression than when given it was given at Home, and drug history had no effect on amphetamine-induced c-fos mRNA expression. Amphetamine 22-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 12890524-9 2003 However, within the subthalamic nucleus, substantia nigra reticulata, and central nucleus of the amygdala prior experience with amphetamine in the Novel but not Home environment enhanced the effect of an amphetamine challenge injection on c-fos mRNA expression. Amphetamine 128-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 239-244 12890524-9 2003 However, within the subthalamic nucleus, substantia nigra reticulata, and central nucleus of the amygdala prior experience with amphetamine in the Novel but not Home environment enhanced the effect of an amphetamine challenge injection on c-fos mRNA expression. Amphetamine 204-215 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 239-244 12890524-10 2003 In contrast, there was a decrease in c-fos mRNA expression in amphetamine-pretreated animals, regardless of environmental context, in the ventral portion of the far caudal striatum. Amphetamine 62-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 12890524-11 2003 Reexposure to an environment previously paired with amphetamine produced a conditioned increase in c-fos mRNA expression in portions of the caudate-putamen, the subthalamic nucleus, the nucleus accumbens shell and a conditioned decrease in c-fos mRNA expression in the central nucleus of the amygdala. Amphetamine 52-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 12890524-11 2003 Reexposure to an environment previously paired with amphetamine produced a conditioned increase in c-fos mRNA expression in portions of the caudate-putamen, the subthalamic nucleus, the nucleus accumbens shell and a conditioned decrease in c-fos mRNA expression in the central nucleus of the amygdala. Amphetamine 52-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 240-245 12927219-4 2003 The main finding of this study is that cholinergic interneurons located in the shell compartment of the nucleus accumbens and the ventromedial striatum were activated, as measured by Fos labeling, following a 1 h session of the self-administration of cocaine in rats. Cocaine 251-258 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 183-186 12927220-11 2003 In rats treated with intra-CeA saline, WIN55,212-2 reduced the incidence of formalin-induced nociceptive behaviors and also reduced formalin-evoked c-fos expression in both superficial and deep laminae of the spinal cord dorsal horn. Sodium Chloride 31-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 12927220-11 2003 In rats treated with intra-CeA saline, WIN55,212-2 reduced the incidence of formalin-induced nociceptive behaviors and also reduced formalin-evoked c-fos expression in both superficial and deep laminae of the spinal cord dorsal horn. Formaldehyde 132-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 14521986-0 2003 Stimulant doses of caffeine induce c-FOS activation in orexin/hypocretin-containing neurons in rat. Caffeine 19-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 14521986-7 2003 Compared with saline, all doses of caffeine increased the number of cells immunoreactive for both orexin and c-Fos. Caffeine 35-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 14521986-10 2003 In contrast, caffeine significantly increased the number of non-orexin-immunoreactive neurons expressing c-Fos only in the dorsomedial nucleus. Caffeine 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 14522010-5 2003 In situ hybridization maps of c-fos mRNA expression showed that in the intact hemisphere, ampakine cotreatment markedly increased c-fos expression in parietal, sensori-motor neocortex above that found in rats treated with methamphetamine alone. ampakine 90-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 14522010-5 2003 In situ hybridization maps of c-fos mRNA expression showed that in the intact hemisphere, ampakine cotreatment markedly increased c-fos expression in parietal, sensori-motor neocortex above that found in rats treated with methamphetamine alone. ampakine 90-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 14596843-5 2003 Pretreatment with d-Phe CRF in the MRN selectively attenuated the increases in c-fos mRNA induced by footshock in the central nucleus of the amygdala (CeA). D-phenylalanine 18-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 12559087-8 2003 In addition, our studies using the inducible immediate early gene c-fos as a marker of activated neurons demonstrated a significant stress-induced activation in perikarya colocalizing urocortin- and cocaine and amphetamine-regulated transcript-ir in the Edinger-Westphal nucleus. Amphetamine 211-222 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 12617970-0 2003 A peripheral cannabinoid mechanism suppresses spinal fos protein expression and pain behavior in a rat model of inflammation. Cannabinoids 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 12617970-6 2003 In immunocytochemical studies, WIN55,212-2 suppressed the development of carrageenan-evoked Fos protein expression in the lumbar dorsal horn of the spinal cord relative to vehicle treatment. Carrageenan 73-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 12617970-8 2003 The suppressive effects of WIN55,212-2 (30 microg intraplantarly) on carrageenan-evoked Fos protein expression and pain behavior were blocked by local administration of either the CB(2) antagonist SR144528 (30 microg intraplantarly) or the CB(1) antagonist SR141716A (100 microg intraplantarly). win55 27-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 14622917-0 2003 Role of NR2B-containing N-methyl-D-aspartate receptors in haloperidol-induced c-Fos expression in the striatum and nucleus accumbens. Haloperidol 58-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 14622917-1 2003 Administration of haloperidol in rats leads to a robust induction of immediate-early genes including c-Fos throughout the striatum, which is significantly attenuated by pretreatment with the non-competitive N-methyl-D-aspartate (NMDA) receptor antagonist, MK-801. Haloperidol 18-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 14622917-1 2003 Administration of haloperidol in rats leads to a robust induction of immediate-early genes including c-Fos throughout the striatum, which is significantly attenuated by pretreatment with the non-competitive N-methyl-D-aspartate (NMDA) receptor antagonist, MK-801. N-Methylaspartate 207-227 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 14622917-1 2003 Administration of haloperidol in rats leads to a robust induction of immediate-early genes including c-Fos throughout the striatum, which is significantly attenuated by pretreatment with the non-competitive N-methyl-D-aspartate (NMDA) receptor antagonist, MK-801. Dizocilpine Maleate 256-262 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 14622917-4 2003 Furthermore, to determine whether NMDA receptor subtype dependence of haloperidol-induced c-Fos expression is unique to the binding profile of haloperidol or whether it is a property of D2 receptor antagonism, the selective D2/D3 dopamine receptor antagonist, raclopride, was also used. Haloperidol 70-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 14622917-6 2003 In the medial part of the striatum, this attenuation was almost as marked as that seen following pretreatment with MK-801; however, in the lateral part MK-801 pretreatment led to a significantly greater reduction in the number of c-Fos positive nuclei than did Ro 25-6981 pretreatment. Dizocilpine Maleate 152-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 230-235 14622917-7 2003 This suggests that NR2B-containing NMDA receptors are involved in mediating most of the NMDA-dependent c-Fos expression in the medial striatum, but only responsible for mediating part of this induction in the lateral striatum. N-Methylaspartate 35-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 14622917-8 2003 Furthermore, the pattern of attenuation of raclopride-induced c-Fos expression following Ro 25-6981 pretreatment was similar to that of haloperidol-induced c-Fos expression, indicating that the NMDA receptor subtype dependence of haloperidol-induced c-Fos expression is a property of D2 antagonism. Raclopride 43-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 14622917-8 2003 Furthermore, the pattern of attenuation of raclopride-induced c-Fos expression following Ro 25-6981 pretreatment was similar to that of haloperidol-induced c-Fos expression, indicating that the NMDA receptor subtype dependence of haloperidol-induced c-Fos expression is a property of D2 antagonism. Haloperidol 136-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 14622917-8 2003 Furthermore, the pattern of attenuation of raclopride-induced c-Fos expression following Ro 25-6981 pretreatment was similar to that of haloperidol-induced c-Fos expression, indicating that the NMDA receptor subtype dependence of haloperidol-induced c-Fos expression is a property of D2 antagonism. Haloperidol 136-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 14622917-9 2003 The results indicate that NR2B-containing NMDA receptors are mainly involved in mediating haloperidol-induced c-Fos expression in the medial or "limbic" striatum, and suggest that NR2A-containing NMDA receptors may preferentially mediate haloperidol induced c-Fos expression in the lateral or "motor" striatum. Haloperidol 90-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 14622917-9 2003 The results indicate that NR2B-containing NMDA receptors are mainly involved in mediating haloperidol-induced c-Fos expression in the medial or "limbic" striatum, and suggest that NR2A-containing NMDA receptors may preferentially mediate haloperidol induced c-Fos expression in the lateral or "motor" striatum. Haloperidol 90-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 258-263 12809695-8 2003 AM1241 suppressed carrageenan-evoked Fos protein expression in the superficial and neck region of the dorsal horn but not in the nucleus proprius or the ventral horn. AM 1241 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 12809695-8 2003 AM1241 suppressed carrageenan-evoked Fos protein expression in the superficial and neck region of the dorsal horn but not in the nucleus proprius or the ventral horn. Carrageenan 18-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 12809695-9 2003 The suppression of carrageenan-evoked Fos protein expression induced by AM1241 was blocked by coadministration of SR144528 in all spinal laminae. Carrageenan 19-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 12809695-9 2003 The suppression of carrageenan-evoked Fos protein expression induced by AM1241 was blocked by coadministration of SR144528 in all spinal laminae. SR 144528 114-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 12809699-0 2003 Morphine induction of c-fos expression in the rat forebrain through glutamatergic mechanisms: role of non-n-methyl-D-aspartate receptors. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 12809699-1 2003 Acute injection of morphine induces expression of the immediate-early genes c-Fos and JunB in several forebrain regions of the rat, in part through an N-methyl-D-aspartate (NMDA) receptor-dependent mechanism. Morphine 19-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 12809699-7 2003 Using double immunocytochemistry, we found that morphine induced c-Fos in neurons containing the GluR2/3, but not the GluR1 and rarely the GluR4, subunits of the AMPA receptor. Morphine 48-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 12809699-13 2003 Pretreatment with AIDA completely blocked morphine-induced c-Fos expression in the caudate-putamen.Taken together, these results demonstrate involvement of both AMPA and type I metabotropic glutamate receptors in the acute effects of morphine on the forebrain, supporting an important role for glutamatergic neurotransmission mediated by non-NMDA glutamate receptors in morphine"s actions. Morphine 42-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 12809699-13 2003 Pretreatment with AIDA completely blocked morphine-induced c-Fos expression in the caudate-putamen.Taken together, these results demonstrate involvement of both AMPA and type I metabotropic glutamate receptors in the acute effects of morphine on the forebrain, supporting an important role for glutamatergic neurotransmission mediated by non-NMDA glutamate receptors in morphine"s actions. Morphine 234-242 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 12809699-13 2003 Pretreatment with AIDA completely blocked morphine-induced c-Fos expression in the caudate-putamen.Taken together, these results demonstrate involvement of both AMPA and type I metabotropic glutamate receptors in the acute effects of morphine on the forebrain, supporting an important role for glutamatergic neurotransmission mediated by non-NMDA glutamate receptors in morphine"s actions. Morphine 234-242 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 12831851-2 2003 Subarachnoid administration of adenosine (AD) or AD agonists promotes sleep and induces expression of Fos protein in VLPO neurons. Adenosine 31-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 12831851-2 2003 Subarachnoid administration of adenosine (AD) or AD agonists promotes sleep and induces expression of Fos protein in VLPO neurons. Adenosine 42-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 12831870-7 2003 In addition, Fos-immunostaining induced by SKF-82958 in caudate-putamen (CPu) and nucleus accumbens (Nac) was greater in food-restricted than ad libitum-fed rats, as was staining induced by quinpirole in globus pallidus and ventral pallidum. Quinpirole 190-200 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 12831870-11 2003 Finally, food restriction increased quinpirole-stimulated [(35)S]guanosine triphosphate-gammaS binding in CPu, suggesting that increased functional coupling between D2 DA receptors and G(i) may account for the augmented behavioral and pallidal c-Fos responses to quinpirole. Quinpirole 36-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 244-249 12617970-8 2003 The suppressive effects of WIN55,212-2 (30 microg intraplantarly) on carrageenan-evoked Fos protein expression and pain behavior were blocked by local administration of either the CB(2) antagonist SR144528 (30 microg intraplantarly) or the CB(1) antagonist SR141716A (100 microg intraplantarly). Carrageenan 69-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 12617970-8 2003 The suppressive effects of WIN55,212-2 (30 microg intraplantarly) on carrageenan-evoked Fos protein expression and pain behavior were blocked by local administration of either the CB(2) antagonist SR144528 (30 microg intraplantarly) or the CB(1) antagonist SR141716A (100 microg intraplantarly). SR 144528 197-205 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 12507695-9 2003 It also significantly attenuated the increase of Fos-Ir neurons observed in ipsilateral TNC laminae I-II after formalin injection. Formaldehyde 111-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 12507695-10 2003 If the proper VNS effect on Fos-expression was subtracted, the reduction of formalin-induced nociceptor activation was 55%. Formaldehyde 76-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 12445834-0 2003 Chronic treatment with the antidepressant tianeptine attenuates lipopolysaccharide-induced Fos expression in the rat paraventricular nucleus and HPA axis activation. tianeptine 42-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-94 12669586-0 2003 [Dizocilpine inhibits suppression of c-fos gene by delta-sleep peptide in the rat hypothalamic paraventricular nucleus]. Dizocilpine Maleate 1-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 12669586-1 2003 The effects of intracerebroventricular administration of the non-competitive NMDA-receptor blocker Dizocilpine (MK-801) on delta sleep-inducing peptide (DSIP) suppression of c-fos induction, were studied. Dizocilpine Maleate 99-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 174-179 12669586-1 2003 The effects of intracerebroventricular administration of the non-competitive NMDA-receptor blocker Dizocilpine (MK-801) on delta sleep-inducing peptide (DSIP) suppression of c-fos induction, were studied. Dizocilpine Maleate 112-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 174-179 12669586-3 2003 MK-801 injection inhibits immediate early gene c-fos suppression by DSIP in the parvocellular paraventricular hypothalamus. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 12422375-4 2003 Acute serotonergic lesions with p-chlorophenylalanine suppressed the expression of Fos induced by 1 mg/kg of amphetamine in both the grafted and the contralateral striatum. Fenclonine 32-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 12422375-4 2003 Acute serotonergic lesions with p-chlorophenylalanine suppressed the expression of Fos induced by 1 mg/kg of amphetamine in both the grafted and the contralateral striatum. Amphetamine 109-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 12422375-5 2003 Chronic serotonergic denervation with 5,7-dihydroxytryptamine induced a significant reduction in Fos expression in both the grafted and nongrafted striata and a nonsignificant reduction in the contraversive rotation. 5,7-Dihydroxytryptamine 38-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 12422375-6 2003 In DA-innervated striata, Prazosin significantly reduced the expression of Fos but only in the presence of serotonergic innervation. Prazosin 26-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 12470860-0 2002 The hallucinogen d-lysergic acid diethylamide (d-LSD) induces the immediate-early gene c-Fos in rat forebrain. Lysergic Acid Diethylamide 17-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 12470860-0 2002 The hallucinogen d-lysergic acid diethylamide (d-LSD) induces the immediate-early gene c-Fos in rat forebrain. Lysergic Acid Diethylamide 47-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 12470860-3 2002 ); c-Fos protein expression was assessed at 30 min, and 1, 2 and 4 h following d-LSD injection. Deuterium 39-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 3-8 12470868-0 2002 The effects of sympathectomy on capsaicin-evoked fos expression of spinal dorsal horn GABAergic neurons. Capsaicin 32-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 12470868-3 2002 We have recently reported that NMDA or non-NMDA antagonists by intrathecal pretreatment attenuate the increased Fos expression in spinal dorsal horn GABAergic neurons after intradermal injection of CAP in rats. N-Methylaspartate 31-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 12470868-3 2002 We have recently reported that NMDA or non-NMDA antagonists by intrathecal pretreatment attenuate the increased Fos expression in spinal dorsal horn GABAergic neurons after intradermal injection of CAP in rats. N-Methylaspartate 43-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 12464353-2 2002 Upregulated preproendothelin-1, endothelin ET(A) receptor, TGF-beta(1), c-fos, and type I collagen expression and extracellular signal-regulated kinase activities were suppressed by celiprolol. Celiprolol 182-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 12464353-4 2002 These observations suggested that extracellular signal-regulated kinase and c-fos gene pathway may contribute to the cardiovascular remodeling of DOCA rats, and that cardioprotective effects of celiprolol on cardiovascular remodeling may be mediated, at least in part, by suppressed expression of endothelin-1 and TGF-beta(1). Desoxycorticosterone Acetate 146-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 12499850-0 2002 Rat strain differences to fluoxetine in striatal Fos-like proteins. Fluoxetine 26-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 12499850-3 2002 We examined the effects of fluoxetine on the induction of Fos-like proteins in Long-Evans and Sprague-Dawley rat brains. Fluoxetine 27-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 12499850-4 2002 Fluoxetine elicited a strong induction of Fos in the striatum of Long-Evans but not Sprague-Dawley rats following acute drug exposure. Fluoxetine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 12499850-6 2002 These differences in Fos expression between rat strains may represent variability in post-receptor pathways that ultimately mediate the therapeutic actions of fluoxetine. Fluoxetine 159-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 12431774-5 2002 Formalin injections caused an increase in Fos expression as well as a release of SP, but not Endo2 from the ipsilateral side dorsal horn in L4-5. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 12456428-0 2002 Isoflurane, but not halothane, depresses c-fos expression in rat spinal cord at concentrations that suppress reflex movement after supramaximal noxious stimulation. Isoflurane 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 12459672-1 2002 BACKGROUND: In a previous study, the authors found that nitrous oxide (N2O) exposure induces c-Fos (an immunohistochemical marker of neuronal activation) in spinal cord gamma-aminobutyric acid-mediated (GABAergic) neurons in Fischer rats. Nitrous Oxide 56-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 12459672-1 2002 BACKGROUND: In a previous study, the authors found that nitrous oxide (N2O) exposure induces c-Fos (an immunohistochemical marker of neuronal activation) in spinal cord gamma-aminobutyric acid-mediated (GABAergic) neurons in Fischer rats. Nitrous Oxide 71-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 12459672-1 2002 BACKGROUND: In a previous study, the authors found that nitrous oxide (N2O) exposure induces c-Fos (an immunohistochemical marker of neuronal activation) in spinal cord gamma-aminobutyric acid-mediated (GABAergic) neurons in Fischer rats. gamma-Aminobutyric Acid 169-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 12459672-7 2002 N2O exposure induced c-Fos expression in the spinal cord, which was blocked by prazosin and naloxone but not by other drugs. Nitrous Oxide 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 12459672-7 2002 N2O exposure induced c-Fos expression in the spinal cord, which was blocked by prazosin and naloxone but not by other drugs. Prazosin 79-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 12459672-7 2002 N2O exposure induced c-Fos expression in the spinal cord, which was blocked by prazosin and naloxone but not by other drugs. Naloxone 92-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 12459672-8 2002 N2O-induced c-Fos expression was colocalized with alpha1 adrenoceptor immunoreactivity in laminae III-IV. Nitrous Oxide 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 12445576-0 2002 Effect of 5-HT1A receptor-mediated serotonin augmentation on Fos immunoreactivity in rat brain. Serotonin 35-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 12429561-0 2002 Estradiol treatment increases CCK-induced c-Fos expression in the brains of ovariectomized rats. Estradiol 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 12473086-5 2002 A neurotoxin, 5-amino-2,4-dihydroxy-alpha-methylphenylethylamine, microinjected into the vicinity of the supraoptic nucleus, selectively depleted the noradrenaline contents of the nucleus and blocked the Fos expression in the supraoptic nucleus after the unconditioned or conditioned fear stimulus. 5-amino-2,4-dihydroxy-alpha-methylphenylethylamine 14-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 204-207 12492441-3 2002 In order to distinguish between these possibilities, we studied amphetamine-induced c-fos immunoreactivity in subregions of rat striatum (patch and matrix compartments of caudate-putamen and nucleus accumbens core and shell) in drug-naive rats, as well as during long-term expression of amphetamine sensitization. Amphetamine 64-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 12492441-4 2002 We found that, in sensitized animals, amphetamine (1.0 mg/kg) evoked an increase in the ratio of c-fos-immunopositive cells in striatal patch and matrix compartments, suggesting a preferential involvement of striatal patches in the sensitized response to amphetamine. Amphetamine 38-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 12492441-5 2002 In drug-naive rats, amphetamine (0.5-5.0 mg/kg) dose-dependently increased c-fos expression in all striatal subregions. Amphetamine 20-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 12492441-6 2002 Remarkably, the highest dose of amphetamine also evoked an increase in patch : matrix ratio of c-fos immunoreactivity. Amphetamine 32-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 12492441-7 2002 In nucleus accumbens core and shell of amphetamine- and saline-pretreated animals, amphetamine (1.0 mg/kg) evoked comparable increases in c-fos expression. Amphetamine 39-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 12492441-7 2002 In nucleus accumbens core and shell of amphetamine- and saline-pretreated animals, amphetamine (1.0 mg/kg) evoked comparable increases in c-fos expression. Sodium Chloride 56-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 12492441-7 2002 In nucleus accumbens core and shell of amphetamine- and saline-pretreated animals, amphetamine (1.0 mg/kg) evoked comparable increases in c-fos expression. Amphetamine 83-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 12492441-9 2002 In addition, they suggest that the shift in amphetamine-induced c-fos expression from striatal matrix to patches in sensitized animals is the consequence of a change in the sensitivity to amphetamine, rather than a long-term circuitry reorganization that is exclusive to the sensitized state. Amphetamine 44-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 12492441-9 2002 In addition, they suggest that the shift in amphetamine-induced c-fos expression from striatal matrix to patches in sensitized animals is the consequence of a change in the sensitivity to amphetamine, rather than a long-term circuitry reorganization that is exclusive to the sensitized state. Amphetamine 188-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 12454857-4 2002 RESULTS: Indomethacin-induced ileitis in Lewis rats leads to specific reductions in ileal ASBT messenger RNA and protein levels, whereas c-jun and c-fos are induced. Indomethacin 9-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-152 12454857-7 2002 Indomethacin (in vivo) or proinflammatory cytokine (in vitro) treatment leads to serine phosphorylation and nuclear translocation of c-fos. Indomethacin 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 12506852-2 2002 We demonstrated previously that a long-duration restraint stress (RTS) evoked adaptive change, characterized by transient increase and gradual recovery to basal level in c-fos mRNA/c-fos protein (Fos) expression in the hypothalamic paraventricular nucleus (PVN) and in plasma adrenocorticotropin (ACTH) levels, although circulating corticosterone (CORT) remained at a high level. Corticosterone 332-346 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-175 12506852-2 2002 We demonstrated previously that a long-duration restraint stress (RTS) evoked adaptive change, characterized by transient increase and gradual recovery to basal level in c-fos mRNA/c-fos protein (Fos) expression in the hypothalamic paraventricular nucleus (PVN) and in plasma adrenocorticotropin (ACTH) levels, although circulating corticosterone (CORT) remained at a high level. Corticosterone 332-346 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 196-199 12506852-4 2002 Superimposed CS-induced re-expression of c-fos mRNA/Fos in the parvocellular region of the PVN (PVNpv) was observed in 16 hours RTS rats. Cesium 13-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 12506852-4 2002 Superimposed CS-induced re-expression of c-fos mRNA/Fos in the parvocellular region of the PVN (PVNpv) was observed in 16 hours RTS rats. Cesium 13-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 12506852-8 2002 The present study indicated that c-fos gene expression in PVN induced by superimposed CS was normo-responsive, ACTH secretion was hyper-responsive (facilitation), CORT secretion was slightly increased, but not hyper-responsive in long-duration RTS rats. Cesium 86-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 12489068-0 2002 Chronic alcohol feeding and its influence on c-Fos and heat shock protein-70 gene expression in different brain regions of male and female rats. Alcohols 8-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 12504594-3 2002 We show here that nicotine can alter gene expression in rat hippocampal neurons, as reflected by activation of the transcription factor CREB and appearance of the immediate early gene product c-Fos. Nicotine 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 192-197 12445632-6 2002 On the other hand, c-Fos expression induced by BmK venom could be suppressed partially by systemic morphine in a dose-dependent manner. Morphine 99-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 12464447-10 2002 The c-fos gene expression was potently induced by haloperidol in caudate-putamen and nucleus accumbens, and by clozapine only in the accumbens. Haloperidol 50-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 12464447-10 2002 The c-fos gene expression was potently induced by haloperidol in caudate-putamen and nucleus accumbens, and by clozapine only in the accumbens. Clozapine 111-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 12464447-11 2002 The adjunction of D-cycloserine enhanced c-fos expression only for clozapine in both regions of the forebrain. Cycloserine 18-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 12464447-11 2002 The adjunction of D-cycloserine enhanced c-fos expression only for clozapine in both regions of the forebrain. Clozapine 67-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 12607946-0 2002 Increased immunoreactivity of POMC-derived neuropeptides and immediate-early gene-derived proteins (c-Fos and Egr-1 proteins) as an early step of acute cocaine-induced stressor effects: comparison with the effects of immobilization stress. Cocaine 152-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 12445576-1 2002 The consequences of pharmacologically evoked augmented serotonin (5-hydroxytryptamine, 5-HT) release on neuronal activity in the brain, as reflected by the cellular expression of the immediate early gene c-fos, were studied. Serotonin 55-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 204-209 12445576-1 2002 The consequences of pharmacologically evoked augmented serotonin (5-hydroxytryptamine, 5-HT) release on neuronal activity in the brain, as reflected by the cellular expression of the immediate early gene c-fos, were studied. Serotonin 87-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 204-209 12445695-2 2002 After a standard PAP immunocytochemical protocol, Fos-like immunoreactivity was observed in the paraventricular nucleus (PVN), supraoptic nucleus (SON), median preoptic area (MnPO), anterior hypothalamus (AH) and posterior hypothalamus (PH) of control (vehicle; n=6) and diabetic rats (Sprague-Dawley rats injected with STZ 65 mg/kg/ip 4 weeks prior to the experiment; n=6). Streptozocin 320-323 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 12419523-8 2002 Naltrexone injections into morphine-dependent rats caused a dramatic increase in c-fos as compared to control rats. Naltrexone 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 12419523-8 2002 Naltrexone injections into morphine-dependent rats caused a dramatic increase in c-fos as compared to control rats. Morphine 27-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 12429408-0 2002 Locomotor sensitization to cocaine is associated with increased Fos expression in the accumbens, but not in the caudate. Cocaine 27-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 12200434-9 2002 Moreover, glucose stimulates the transactivation functions of c-Fos and USF1, but not c-Jun, in one-hybrid assays. Glucose 10-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 12429408-3 2002 In the present experiment, we compared the ability of increasing doses of cocaine to induce Fos in the accumbens and caudate of rats following a treatment procedure (7 once daily injections of 15 mg/kg of cocaine or the saline vehicle) shown to produce robust and persistent (1 week) locomotor sensitization. Cocaine 74-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 12429408-4 2002 In sensitized animals, there was a leftward shift in the dose-response curve for cocaine induction of Fos in the accumbens, but not in the caudate. Cocaine 81-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 12429408-5 2002 These results provide the first parametric evidence for sensitization of cocaine-induced Fos expression in the accumbens. Cocaine 73-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 12372804-1 2002 cAMP/PKA signaling transiently stimulates mRNA expression of immediate-early genes, including IL-6 and c-fos. Cyclic AMP 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 12401615-5 2002 Although the number of c-fos-positive cells in the lumbar spinal cord in the nitrous oxide group was not decreased, that in the xenon group decreased. Nitrous Oxide 77-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 12401615-7 2002 Inhaled xenon suppressed nociceptive behaviors, c-fos expression, and activation of the NMDA receptor during the formalin test in rats. Xenon 8-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 12401615-9 2002 IMPLICATIONS: Inhaled xenon suppressed nociceptive behaviors, c-fos expression, and activation of the N-methyl-D-aspartate receptor during the formalin test in rats. Xenon 22-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 12393773-3 2002 METHODS: We used an in vivo rat model of brain injury in which N-methyl-DL-aspartic acid (NMA) is injected subcutaneously (s.c.) and c-Fos expression in the arcuate nucleus is used as a measure of injury. N-methyl-DL-aspartic acid 90-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 12393773-5 2002 RESULTS: Xenon dose-dependently suppressed NMA-induced c-Fos expression in the arcuate nucleus with an IC(50) of 47 (2)% atm. Xenon 9-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 12393773-5 2002 RESULTS: Xenon dose-dependently suppressed NMA-induced c-Fos expression in the arcuate nucleus with an IC(50) of 47 (2)% atm. N-methyl-DL-aspartic acid 43-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 12393773-7 2002 Both nitrous oxide and ketamine dose-dependently increased c-Fos expression in PC/RS cortices; in contrast, xenon produced no significant effect. Nitrous Oxide 5-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 12393773-7 2002 Both nitrous oxide and ketamine dose-dependently increased c-Fos expression in PC/RS cortices; in contrast, xenon produced no significant effect. Ketamine 23-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 12390534-5 2002 In contrast, chlomethiazole and SB203580 potently inhibited the IL-1beta-induced expression of c-fos and inducible nitric oxide synthase, as monitored by northern blot and quantitative RT-PCR, respectively. chlomethiazole 13-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 12390534-5 2002 In contrast, chlomethiazole and SB203580 potently inhibited the IL-1beta-induced expression of c-fos and inducible nitric oxide synthase, as monitored by northern blot and quantitative RT-PCR, respectively. SB 203580 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 12390534-6 2002 Because IL-1beta-induced expression of c-fos and inducible nitric oxide synthase is believed to directly contribute to the pathology of cerebral ischaemic injury, the results suggest a direct mechanism for the neuroprotective effects of chlomethiazole and SB203580, and further establish the anti-inflammatory properties of chlomethiazole. chlomethiazole 237-251 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 12390534-6 2002 Because IL-1beta-induced expression of c-fos and inducible nitric oxide synthase is believed to directly contribute to the pathology of cerebral ischaemic injury, the results suggest a direct mechanism for the neuroprotective effects of chlomethiazole and SB203580, and further establish the anti-inflammatory properties of chlomethiazole. SB 203580 256-264 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 12390534-6 2002 Because IL-1beta-induced expression of c-fos and inducible nitric oxide synthase is believed to directly contribute to the pathology of cerebral ischaemic injury, the results suggest a direct mechanism for the neuroprotective effects of chlomethiazole and SB203580, and further establish the anti-inflammatory properties of chlomethiazole. chlomethiazole 324-338 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 12499582-4 2002 The c-fos mRNA levels returned to control levels 6 h after the naloxone challenge. Naloxone 63-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 12499582-7 2002 These results suggest that physical dependence develops in infant rats following repeated morphine administration and that the increment of c-fos mRNA levels is a useful indicator for naloxone-precipitated morphine withdrawal in infant as well as in adult rats. Naloxone 184-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 12499582-7 2002 These results suggest that physical dependence develops in infant rats following repeated morphine administration and that the increment of c-fos mRNA levels is a useful indicator for naloxone-precipitated morphine withdrawal in infant as well as in adult rats. Morphine 206-214 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 12431845-2 2002 We investigated the mechanism of action of atomoxetine in ADHD by evaluating the interaction of atomoxetine with monoamine transporters, the effects on extracellular levels of monoamines, and the expression of the neuronal activity marker Fos in brain regions. Atomoxetine Hydrochloride 43-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 239-242 12491779-7 2002 The number of Fos-positive neurons in the CA1 hippocampus demonstrated a statistically significant negative correlation with the number of working memory errors, suggesting that the DHA-induced improvement in spatial cognition is associated with increased Fos expression in the CA1 hippocampus. Docosahexaenoic Acids 182-185 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 12491779-7 2002 The number of Fos-positive neurons in the CA1 hippocampus demonstrated a statistically significant negative correlation with the number of working memory errors, suggesting that the DHA-induced improvement in spatial cognition is associated with increased Fos expression in the CA1 hippocampus. Docosahexaenoic Acids 182-185 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 256-259 12587265-0 2002 [C-fos gene expression in the rat spinal cord and brain cells during stress and the use of different types of halothane anesthesia]. Halothane 110-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 1-6 12587265-2 2002 Using of 1.5% light halothane narcosis allowed the detection of c-Fos-like proteins expression in the spinal cord cells only. Halothane 20-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 12587265-3 2002 Under initial 1.5% halothane narcosis, c-Fos-like proteins expression in the rat spinal cord (lumbar segments) and the brain cells was observed after placing the rats into the hammock, noxious mechanical stimulation (NMS) or high frequency electromagnetic irradiation of the skin (EHF). Halothane 19-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 12587265-6 2002 EHF irradiation of the skin decreased the intensity of c-Fos-like proteins synthesis induced by NMS in the most of the investigated structures (LHA, VMH, DMH and AHA by 32.8, 29, 15 and 33%, resp.). Dimenhydrinate 154-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 12211088-5 2002 The degrees of Fos activation that occurred in GABAergic cells and cells immunopositive for NMDA receptors were similar in the DCIC, CIC, ECIC, and DpG following AS. CHVP protocol 128-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-18 12211088-5 2002 The degrees of Fos activation that occurred in GABAergic cells and cells immunopositive for NMDA receptors were similar in the DCIC, CIC, ECIC, and DpG following AS. dpg 148-151 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-18 12385797-5 2002 Essential oil increased the c-Fos expression in the arcuate n. of the hypothalamus. Oils, Volatile 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 12431845-7 2002 The expression of the neuronal activity marker Fos was increased 3.7-fold in PFC by atomoxetine administration, but was not increased in the striatum or nucleus accumbens, consistent with the regional distribution of increased DA(EX). Atomoxetine Hydrochloride 84-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 12372551-4 2002 Exposure to a light pulse (LP) 1 h prior to sacrifice led to increased Fos expression in subjects maintained for 2 weeks in constant gravity (either at approximately 0 or 1 G). leucylproline 27-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 12385797-6 2002 Essential oil and formalin increased c-Fos in the paraventricular n. of the hypothalamus and in the dentate gyrus of the hippocampus. Oils, Volatile 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 12385797-6 2002 Essential oil and formalin increased c-Fos in the paraventricular n. of the hypothalamus and in the dentate gyrus of the hippocampus. Formaldehyde 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 12385797-7 2002 In the paraventricular n. of the thalamus formalin induced higher c-Fos than control in both sexes; when formalin treatment was carried out in presence of essential oil, c-Fos further increased in males, but remained at control levels in females. Formaldehyde 42-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 12385797-7 2002 In the paraventricular n. of the thalamus formalin induced higher c-Fos than control in both sexes; when formalin treatment was carried out in presence of essential oil, c-Fos further increased in males, but remained at control levels in females. Formaldehyde 105-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-175 12385797-7 2002 In the paraventricular n. of the thalamus formalin induced higher c-Fos than control in both sexes; when formalin treatment was carried out in presence of essential oil, c-Fos further increased in males, but remained at control levels in females. Oils, Volatile 155-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-175 12372551-5 2002 Within 24 h of a gravitational change (launch or landing), the Fos response to LP was abolished. leucylproline 79-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 12270498-0 2002 Subfornical organ disconnection and Fos-like immunoreactivity in hypothalamic nuclei after intragastric hypertonic saline. Sodium Chloride 115-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-39 12228050-7 2002 In the virgin rats, and in the pregnant rats treated with L-NAME, atrial distension significantly increased hypothalamic c-fos expression. NG-Nitroarginine Methyl Ester 58-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-126 12228057-7 2002 Moreover, left adrenalectomy induced c-Fos immunolabeling in ipsilateral dorsal spinal cord that was prevented by capsaicin treatment. Capsaicin 114-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 12270498-2 2002 However, in response to mild doses of hypertonic saline, Fos-like immunoreactivity (Fos-ir) is absent in the SFO whereas it is well expressed in the hypothalamic supraoptic (SON) and paraventricular (PVN) nuclei. Sodium Chloride 49-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 12270498-2 2002 However, in response to mild doses of hypertonic saline, Fos-like immunoreactivity (Fos-ir) is absent in the SFO whereas it is well expressed in the hypothalamic supraoptic (SON) and paraventricular (PVN) nuclei. Sodium Chloride 49-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 12270498-7 2002 Thus, the connectivity between SFO and the hypothalamus is critical for the full expression of Fos-ir in the hypothalamus during moderate ig hypertonic saline loading even when the SFO itself does not yet express Fos-ir. Sodium Chloride 152-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 12270498-7 2002 Thus, the connectivity between SFO and the hypothalamus is critical for the full expression of Fos-ir in the hypothalamus during moderate ig hypertonic saline loading even when the SFO itself does not yet express Fos-ir. Sodium Chloride 152-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-216 12244091-5 2002 Prior treatment with chronic intermittent cocaine induced motor sensitization and significantly potentiated the striatal expression of Fos-family early genes in response to stimulation of the motor cortex. Cocaine 42-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-138 12239109-0 2002 Expression of estrogen receptor-alpha and cFos in norepinephrine and epinephrine neurons of young and middle-aged rats during the steroid-induced luteinizing hormone surge. Norepinephrine 50-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-46 12239109-3 2002 The aims of the present study were to determine whether the NE and epinephrine neurons continue to express estrogen receptor (ER)-alpha in middle-aged rats; temporal expression of ER-alpha and cFos changes with age during the steroid-induced surge; and tyrosine hydroxylase (TH), dopamine-beta-hydroxylase (DBH), and phenylethanol-N-methyltransferase mRNA content in catecholaminergic neurons of the brain stem changes during the surge with age. Steroids 226-233 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-197 12239109-8 2002 It is concluded that the reduced catecholamine release during the surge in middle-aged rats is caused, in part, by an altered sensitivity of the NE neurons to estradiol, which results in an aberrant cFos expression and probably not by major deficits in the expression of transmitter synthesizing enzymes or steroid receptors. Catecholamines 33-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-203 12239109-8 2002 It is concluded that the reduced catecholamine release during the surge in middle-aged rats is caused, in part, by an altered sensitivity of the NE neurons to estradiol, which results in an aberrant cFos expression and probably not by major deficits in the expression of transmitter synthesizing enzymes or steroid receptors. Estradiol 159-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-203 12405997-4 2002 Using in situ hybridization, we have analysed in the brain of morphine-dependent rats the effects of acute withdrawal syndrome precipitated by increasing naloxone doses on c-fos mRNA expression. Morphine 62-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-177 12405997-4 2002 Using in situ hybridization, we have analysed in the brain of morphine-dependent rats the effects of acute withdrawal syndrome precipitated by increasing naloxone doses on c-fos mRNA expression. Naloxone 154-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-177 12405997-6 2002 Our mapping study revealed a dissociation between a set of brain structures (extended amygdala, lateral septal nucleus, basolateral amygdala and field CA1 of the hippocampus) which exhibited c-fos mRNA dose-dependent variations from the lowest naloxone doses, and many other structures (dopaminergic and noradrenergic nuclei, motor striatal areas, hypothalamic nuclei and periaqueductal grey) which were less sensitive and recruited only by the higher doses. Naloxone 244-252 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 191-196 12405997-7 2002 In addition, we found opposite dose-dependent variations of c-fos gene expression within the central (increase) and the basolateral (decrease) amygdala after acute morphine withdrawal. Morphine 164-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 12358736-0 2002 Morphine withdrawal-induced c-fos expression in the hypothalamic paraventricular nucleus is dependent on the activation of catecholaminergic neurones. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 12358736-10 2002 Similarly, pre-treatment with the beta antagonist, propranolol (3 mg/kg intraperitoneally), significantly prevented withdrawal-induced Fos expression. Propranolol 51-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-138 12372001-6 2002 Fos expression (an indicator of neuronal activation) in the arcuate nucleus was colocalized in beta-endorphin neurones in both proestrus and parturient rats, but the number of positive cells did not increase during parturition, suggesting lack of activation of beta-endorphin neurones at birth. (2-benzoylethyl)trimethylammonium 95-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 12195434-4 2002 Using c-Fos as a marker of neuronal activation, we identified a possible role for the tuberomammillary nucleus (TMN): when gabazine was microinjected directly into the TMN, it attenuated the sedative response to GABAergic agents. gabazine 123-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 6-11 12500171-1 2002 OBJECTIVES: The content of C-Fos protein was tested in rat pinealocytes in the norm and stress and in case of intranasal administration of Epitalon (Ala-Glu-Asp-Gly), which regulated pineal secretion processes, presumably, via protooncogenes. alanylglutamic acid 149-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 12500171-1 2002 OBJECTIVES: The content of C-Fos protein was tested in rat pinealocytes in the norm and stress and in case of intranasal administration of Epitalon (Ala-Glu-Asp-Gly), which regulated pineal secretion processes, presumably, via protooncogenes. Aspartic Acid 157-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 12500171-1 2002 OBJECTIVES: The content of C-Fos protein was tested in rat pinealocytes in the norm and stress and in case of intranasal administration of Epitalon (Ala-Glu-Asp-Gly), which regulated pineal secretion processes, presumably, via protooncogenes. Glycine 161-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 12377393-6 2002 Intrastriatal blockade of group I mGluRs with N-phenyl-7-(hydroxyimino)cyclopropa[b]chromen-1a-carboxamide (PHCCC; 25 nmol) significantly attenuated amphetamine-induced pCREB, pElk-1, pERK1/2, and Fos immunoreactivity in both medial and lateral areas of the striatum. N-phenyl-7-(hydroxyimino)cyclopropa(b)chromen-1a-carboxamide 46-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 197-200 12377393-12 2002 These data suggest that acute amphetamine is able to facilitate the phosphorylation of CREB, Elk-1, and ERK1/2 signaling proteins and Fos gene expression via a group I mGluR-dependent mechanism in the dorsal striatum. Amphetamine 30-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-137 12392213-0 2002 MK 801 attenuates c-Fos and c-Jun expression after in vitro ischemia in rat neuronal cell cultures but not in PC 12 cells. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 12392213-4 2002 Since activation of glutamate receptors is known to mediate ischemic injury we determined the effect of the noncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist MK 801 on c-Fos and c-Jun expression in both cell culture systems during ischemia. Dizocilpine Maleate 171-177 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 181-186 12392213-10 2002 The addition of MK 801 significantly reduced the expression of c-Fos and c-Jun mRNA in neuronal cultures, whereas no effect was detectable in PC 12 cells. Dizocilpine Maleate 16-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 12370534-0 2002 Regulation of carbachol-induced c-fos mRNA expression in AR42J cells by somatostatin receptor subtypes 1, 2, and 3. Carbachol 14-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 12406515-0 2002 Effect of intrathecal octreotide on thermal hyperalgesia and evoked spinal c-Fos expression in rats with sciatic constriction injury. Octreotide 22-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 12406515-1 2002 This study was designed to determine whether intrathecal octreotide (sandostatin), a synthetic octapeptide derivative of somatostatin, relieved thermal hyperalgesia and reduced the evoked spinal c-Fos expression in rats with chronic constriction injury (CCI) of the sciatic nerve. Octreotide 57-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-200 12406515-1 2002 This study was designed to determine whether intrathecal octreotide (sandostatin), a synthetic octapeptide derivative of somatostatin, relieved thermal hyperalgesia and reduced the evoked spinal c-Fos expression in rats with chronic constriction injury (CCI) of the sciatic nerve. Octreotide 69-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-200 12406529-13 2002 injection of carrageenan, the number of Fos-like immunoreactive (Fos-LI) neurons was significantly increased in all the layers of the ipsilateral spinal cord at L(4-5), with the highest density in laminae I-II and V-VI. Carrageenan 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 12406529-13 2002 injection of carrageenan, the number of Fos-like immunoreactive (Fos-LI) neurons was significantly increased in all the layers of the ipsilateral spinal cord at L(4-5), with the highest density in laminae I-II and V-VI. Carrageenan 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 12406529-14 2002 Intrathecally pre-administered AP5 (10 nmol) or DNQX (100 nmol) significantly reduced the total number of carrageenan-induced Fos-LI neurons. 2-amino-5-phosphopentanoic acid 31-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-129 12406529-14 2002 Intrathecally pre-administered AP5 (10 nmol) or DNQX (100 nmol) significantly reduced the total number of carrageenan-induced Fos-LI neurons. FG 9041 48-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-129 12406529-14 2002 Intrathecally pre-administered AP5 (10 nmol) or DNQX (100 nmol) significantly reduced the total number of carrageenan-induced Fos-LI neurons. Carrageenan 106-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-129 12406529-16 2002 Similarly, following bilateral electroacupuncture stimulation of the "Zu-San-Li" and "Kun-Lun" acupuncture points, the numbers of carrageenan-induced Fos-LI neurons in laminae I-II and V-VI were also markedly reduced. Carrageenan 130-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-153 12406529-17 2002 When a combination of electroacupuncture with 10 nmol AP5 or 100 nmol DNQX was used, the level of Fos expression in the spinal cord induced by carrageenan was significantly lower than electroacupuncture or i.t. 2-amino-5-phosphopentanoic acid 54-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 12406529-17 2002 When a combination of electroacupuncture with 10 nmol AP5 or 100 nmol DNQX was used, the level of Fos expression in the spinal cord induced by carrageenan was significantly lower than electroacupuncture or i.t. FG 9041 70-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 12406529-17 2002 When a combination of electroacupuncture with 10 nmol AP5 or 100 nmol DNQX was used, the level of Fos expression in the spinal cord induced by carrageenan was significantly lower than electroacupuncture or i.t. Carrageenan 143-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 12392621-6 2002 RESULTS: Addition of 0.1 to 50 micromol/L lovastatin into culture medium had no toxicity to hepatic stellate cells, but could significantly inhibit hepatic stellate cell proliferation and provoke G0/G1 phase arrest in dose-dependent manner, and could also markedly inhibit the c-jun and c-fos expression and type IV collagen and laminin secretion, which could partly be antagonized by geranyl geranypyrophosphate. Lovastatin 42-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 287-292 12231231-7 2002 The results showed that mating-with-intromissions induced a significant increase in the percentage of NADPH-d/Fos colabeled neurons in the medial preoptic area (mPOA) and the magnocellular component of the paraventricular nucleus (PVNm) compared to mounts-without-intromission or control treatment. NADP 102-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 12231231-8 2002 Both mating and mounting induced Fos expression in NADPH-d-positive cells in the ventromedial nucleus of hypothalamus (VMN). NADP 51-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 12231231-9 2002 In contrast, the expression of Fos in the NADPH-d-positive neurons in the supraoptic nucleus (SON) and the parvocellular portion of the paraventricular nucleus (PVNp) was not influenced by either mating or mounting although abundant NO-containing neurons were found in the two brain areas. NADP 42-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 12231241-0 2002 Sensitized Fos expression in subterritories of the rat medial prefrontal cortex and nucleus accumbens following amphetamine sensitization as revealed by stereology. Amphetamine 112-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 12231241-3 2002 In the present study, we investigated the effects of amphetamine sensitization on Fos immunoreactivity (Fos-IR) in subterritories of the nucleus accumbens (core and shell) and medial prefrontal cortex (mPFC; dorsal and ventral) using stereology. Amphetamine 53-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 12231241-3 2002 In the present study, we investigated the effects of amphetamine sensitization on Fos immunoreactivity (Fos-IR) in subterritories of the nucleus accumbens (core and shell) and medial prefrontal cortex (mPFC; dorsal and ventral) using stereology. Amphetamine 53-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 12231241-8 2002 Densities of Fos-positive nuclei were enhanced more in the dorsal than the ventral mPFC subterritory, whereas in the nucleus accumbens, densities of Fos-positive nuclei were increased more in the core than the shell of amphetamine-sensitized rats compared to controls. Amphetamine 219-230 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 149-152 12231241-9 2002 These results represent, to our knowledge, the first published report using stereological methods to quantify Fos-IR in the brain and suggest functional specialization of cortical and limbic regions in the expression of behavioral sensitization to amphetamine. Amphetamine 248-259 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 12213641-1 2002 The expression of early response gene proteins c-Fos, c-Jun, and GAP-43 and their association with 6-hydroxydopamine (6-OHDA)-mediated oxidative injury were investigated using catecholaminergic PC12 cell line. Oxidopamine 118-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 12213641-2 2002 Significant induction in the expression of c-Fos (P < 0.01), c-Jun (P < 0.001) and GAP-43 (P < 0.05) was observed following 2 h exposure to 6-OHDA (10(-6) M), which persisted during 24 h of observation. Oxidopamine 140-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 12231642-0 2002 c-Fos expression in ouabain-treated vascular smooth muscle cells from rat aorta: evidence for an intracellular-sodium-mediated, calcium-independent mechanism. Sodium 111-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 12231642-0 2002 c-Fos expression in ouabain-treated vascular smooth muscle cells from rat aorta: evidence for an intracellular-sodium-mediated, calcium-independent mechanism. Calcium 128-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 12231642-2 2002 Incubation of VSMC with ouabain resulted in rapid induction of c-Fos protein expression with an approximately sixfold elevation after 2 h of incubation. Ouabain 24-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 12231642-4 2002 Markedly augmented c-Fos expression was also observed under Na(+)-K(+) pump inhibition in potassium-depleted medium. Potassium 90-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 12231642-9 2002 Augmented c-Fos expression was also observed under VSMC depolarization in high-potassium medium. Potassium 79-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-15 12231642-10 2002 Increments in both c-Fos expression and (45)Ca uptake in depolarized VSMC were abolished under inhibition of L-type Ca(2+) channels with 0.1 microM nicardipine. Nicardipine 148-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 12231642-12 2002 Ouabain-induced c-Fos expression was also insensitive to the presence of nicardipine and [Ca(2+)](o), as well as chelators of [Ca(2+)](o) (EGTA) and [Ca(2+)](i) (BAPTA). Ouabain 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 12231642-12 2002 Ouabain-induced c-Fos expression was also insensitive to the presence of nicardipine and [Ca(2+)](o), as well as chelators of [Ca(2+)](o) (EGTA) and [Ca(2+)](i) (BAPTA). Egtazic Acid 139-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 12223232-3 2002 All drugs produced marked reductions of formalin-induced c-Fos and Zif/268 immunoreactivity in laminae I and II, and laminae V and VI in the rat lumbar spinal cord. Formaldehyde 40-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 12189203-7 2002 Accordingly, a synergistic effect on c-fos expression in striatal sections and on counteracting phencyclidine-induced motor activation was also demonstrated after the central coadministration of A2AR and mGluR5 agonists to rats with intact dopaminergic innervation. Phencyclidine 96-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 12217933-0 2002 Effects of acetaldehyde on c-fos mRNA induction in the paraventricular nucleus following ethanol administration. Acetaldehyde 11-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 12217933-0 2002 Effects of acetaldehyde on c-fos mRNA induction in the paraventricular nucleus following ethanol administration. Ethanol 89-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 12217933-1 2002 AIMS: The effect of acetaldehyde on c-fos mRNA expression in the paraventricular nucleus (PVN) of the rat was examined using in situ hybridization histochemistry. Acetaldehyde 20-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 12217933-5 2002 However, the combination of cyanamide and low dose ethanol resulted in a significant and maximal increase in c-fos mRNA in the PVN. Cyanamide 28-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 12217933-5 2002 However, the combination of cyanamide and low dose ethanol resulted in a significant and maximal increase in c-fos mRNA in the PVN. Ethanol 51-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 12217933-6 2002 High dose ethanol (3 g/kg) resulted in a significant increase in c-fos mRNA. Ethanol 10-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 12217933-8 2002 CONCLUSIONS: These data suggest that acetaldehyde accumulation in blood has an important stimulatory effect on c-fos expression in the PVN at low ethanol concentrations. Acetaldehyde 37-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 12217933-8 2002 CONCLUSIONS: These data suggest that acetaldehyde accumulation in blood has an important stimulatory effect on c-fos expression in the PVN at low ethanol concentrations. Ethanol 146-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 12217933-9 2002 Furthermore, this stimulation of c-fos mRNA appears to be an either/or response: not activated in response to low dose ethanol, but maximally to high dose ethanol. Ethanol 119-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 12217933-9 2002 Furthermore, this stimulation of c-fos mRNA appears to be an either/or response: not activated in response to low dose ethanol, but maximally to high dose ethanol. Ethanol 155-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 12217933-10 2002 These data also provide further evidence for a dissociation between the activation of c-fos and corticotrophin-releasing factor (CRF) mRNA in the PVN, as we have previously demonstrated that this dose of cyanamide alone is sufficient to evoke a sustained increase in plasma corticosterone and an increase in CRF mRNA. Cyanamide 204-213 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 12217933-10 2002 These data also provide further evidence for a dissociation between the activation of c-fos and corticotrophin-releasing factor (CRF) mRNA in the PVN, as we have previously demonstrated that this dose of cyanamide alone is sufficient to evoke a sustained increase in plasma corticosterone and an increase in CRF mRNA. Corticosterone 274-288 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 12351928-6 2002 Adrenal early gene expression of all of the genes studied was strongly induced by the treatment in both rat lines, but the inductions of c-fos, nor1, and nurr1 were significantly lower in the alcohol-sensitive animals. Alcohols 192-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 12169436-4 2002 The numbers of Fos-positive cells in each above-named brain nucleus or in the gastric/duodenal myenteric plexus of insulin-treated rats were negatively correlated with serum glucose levels and significantly increased when glucose levels were lower than 80 mg/dl. Glucose 174-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-18 12169436-4 2002 The numbers of Fos-positive cells in each above-named brain nucleus or in the gastric/duodenal myenteric plexus of insulin-treated rats were negatively correlated with serum glucose levels and significantly increased when glucose levels were lower than 80 mg/dl. Glucose 222-229 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-18 12184990-4 2002 Coadministration of a low dose (1 mg/kg) of CP-99,994 and NMDA receptor antagonist (MK-801), either of which alone did not affect c-fos expression, significantly inhibited c-fos expression at both levels of the spinal cord. cp-99 44-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-177 12184990-4 2002 Coadministration of a low dose (1 mg/kg) of CP-99,994 and NMDA receptor antagonist (MK-801), either of which alone did not affect c-fos expression, significantly inhibited c-fos expression at both levels of the spinal cord. Dizocilpine Maleate 84-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-177 12511984-0 2002 Cycloheximide prevents inhibition of expression of immediate early gene c-fos in paraventricular nuclei of rat hypothalamus produced by delta sleep-inducing peptide. Cycloheximide 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 12511984-1 2002 We studied expression of the immediate early gene c-fos in hypothalamic paraventricular nuclei in rats with different prognostic resistance to emotional stress receiving delta-sleep-inducing peptide after intracerebroventricular administration of cycloheximide. Cycloheximide 247-260 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 12511984-2 2002 Delta-sleep-inducing peptide inhibited expression of the c-fos gene in rats receiving intracerebroventricular injection of physiological saline. Sodium Chloride 137-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 12429227-4 2002 L-DOPA responsiveness was also accompanied by changes at the neuronal level, as shown by changes in the expression of c-fos in the dopamine-depleted striatum. Levodopa 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 12429227-4 2002 L-DOPA responsiveness was also accompanied by changes at the neuronal level, as shown by changes in the expression of c-fos in the dopamine-depleted striatum. Dopamine 131-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 12429227-5 2002 Following 1 week of L-DOPA treatment there was a marked decrease in striatal c-fos expression, compared to single injections, especially evident in the medial and ventral regions and to a lesser extent in the dorsolateral regions of the striatum. Levodopa 20-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 12429227-6 2002 This specific regional expression of c-fos was maintained throughout the 16 weeks of L-DOPA treatment. Levodopa 85-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 12429227-8 2002 Persisting c-fos expression in the dorsolateral striatum might be implicated in the development of dyskinesias when L-DOPA treatment is extended for periods longer than 1 week. Levodopa 116-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 12215476-6 2002 Whereas the re-expression of AT1R at both transcriptional and functional expression levels after baroreceptor activation was discernibly blunted by prior bilateral application into the NTS of an antisense c-fos oligonucleotide (50 pmol), the suppression in SHR was again significantly more intense. Oligonucleotides 211-226 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 205-210 12215476-7 2002 Control pretreatment with the corresponding sense or scrambled c-fos oligonucleotide was ineffective. Oligonucleotides 69-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 12369737-3 2002 Previous immunohistochemical studies using Fos proteins expression as a marker of neuronal activity showed the involvement of arachidonic acid cascade, particularly cyclooxygenase metabolic pathway, in DOI-induced Fos proteins expression in the rat parietal cortex. Arachidonic Acid 126-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 12369737-3 2002 Previous immunohistochemical studies using Fos proteins expression as a marker of neuronal activity showed the involvement of arachidonic acid cascade, particularly cyclooxygenase metabolic pathway, in DOI-induced Fos proteins expression in the rat parietal cortex. Arachidonic Acid 126-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 214-217 12403005-9 2002 The maximum number of c-fos mRNA-positive cells in the anterior field and the paraventricular nucleus of the hypothalamic induced by tetanus toxoid, as compared with reactions to administration of physiological saline, were seen at 6 h. Administration of tetanus toxoid and physiological saline did not active the synthesis of c-fos mRNA in the arcuate or supraoptic nuclei at any time point. Sodium Chloride 211-217 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 12403005-9 2002 The maximum number of c-fos mRNA-positive cells in the anterior field and the paraventricular nucleus of the hypothalamic induced by tetanus toxoid, as compared with reactions to administration of physiological saline, were seen at 6 h. Administration of tetanus toxoid and physiological saline did not active the synthesis of c-fos mRNA in the arcuate or supraoptic nuclei at any time point. Sodium Chloride 288-294 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 12548342-2 2002 Animals treated with the D(1) agonist SKF 38393 showed massive Fos increases in the cerebral cortex, ipsilaterally to the injected striatum, which were counteracted by systemic administration of D(1) antagonist SCH 23390. 1,2,3,4-tetrahydroisoquinoline-7-sulfonamide 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 12548342-3 2002 Conversely, D(2) agonist quinpirole suppressed cortical expression of Fos, while systemic administration of D(2) antagonist eticlopride relieved this blockade. Quinpirole 25-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 12548342-4 2002 As for the basal ganglia, Fos was consistently expressed only in the injected striatum of rats receiving SKF 38393. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 105-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 12112395-9 2002 Similarly, in the second experiment it was found that the D1R-dependent induction by AMPH of Fos, FosB, and JunB, but not NGFI-A, in the NAc was enhanced in rats exposed 1 week earlier to repeated VTA AMPH. Amphetamine 85-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 12112395-9 2002 Similarly, in the second experiment it was found that the D1R-dependent induction by AMPH of Fos, FosB, and JunB, but not NGFI-A, in the NAc was enhanced in rats exposed 1 week earlier to repeated VTA AMPH. vta amph 197-205 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 12147189-7 2002 Midazolam-pretreatment inhibited the increase of Fos protein expression, not cyclic AMP content, in rat spinal cord during morphine withdrawal. Midazolam 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 12177222-5 2002 This potentiation (1) was suppressed by the administration of (R)-alpha-methylhistamine, a H3-receptor agonist, (2) occurred both in the caudate-putamen and nucleus accumbens, and (3) was also observed with a similar pattern on c-fos and neurotensin mRNA expression. alpha-methylhistamine 62-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 228-233 12225868-2 2002 In search for the underlying neuronal mechanisms we investigated the influence of stress on morphine-induced c-fos expression in the brain, and, vice versa, the influence of morphine application on the brain"s c-fos response to stress. Morphine 92-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 12225868-6 2002 The stress-induced c-fos induction was markedly decreased by a moderate (10 mg/kg) dose of morphine. Morphine 91-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 12225868-7 2002 On the other hand, morphine alone (50 mg/kg) caused only a weak c-fos expression in nai;ve animals despite of the rather high dose. Morphine 19-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 12225868-7 2002 On the other hand, morphine alone (50 mg/kg) caused only a weak c-fos expression in nai;ve animals despite of the rather high dose. Sodium Iodide 84-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 12225868-8 2002 If, however, this morphine dose was applied in the presence of a stressful stimulus, a pronounced c-fos expression in the dorsal striatum resulted. Morphine 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 12133574-9 2002 injection of carrageenan, the number of c-Fos-like immunoreactive (c-Fos-LI) neurons was significantly increased in laminae I-II, III-IV and V-VI of the ipsilateral spinal cord at L4-5 with the higher density in laminae I-II and V-VI. Carrageenan 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 12133574-9 2002 injection of carrageenan, the number of c-Fos-like immunoreactive (c-Fos-LI) neurons was significantly increased in laminae I-II, III-IV and V-VI of the ipsilateral spinal cord at L4-5 with the higher density in laminae I-II and V-VI. Carrageenan 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 12133574-11 2002 pre-injected IL-1beta significantly decreased the number of carrageenan-induced c-Fos-LI neurons in laminae I-II in the ipsilateral spinal cord and also inhibited the hyperalgesia induced by i.pl. Carrageenan 60-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 12147189-7 2002 Midazolam-pretreatment inhibited the increase of Fos protein expression, not cyclic AMP content, in rat spinal cord during morphine withdrawal. Morphine 123-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 12145054-0 2002 Midazolam induces expression of c-Fos and EGR-1 by a non-GABAergic mechanism. Midazolam 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 12145054-4 2002 In contrast, midazolam dose- and time-dependently induced expression of c-Fos and EGR-1, which was not affected by antagonists of the benzodiazepine receptors, flumazenil and PK11195. Midazolam 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 12145054-5 2002 The midazolam-induced c-Fos and EGR-1 expression was abolished by PD98059, an inhibitor for mitogen- activated protein kinase/extracellular signal-regulated kinase kinase, suggesting the involvement of extracellular signal-regulated kinases (ERKs). Midazolam 4-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 12145054-5 2002 The midazolam-induced c-Fos and EGR-1 expression was abolished by PD98059, an inhibitor for mitogen- activated protein kinase/extracellular signal-regulated kinase kinase, suggesting the involvement of extracellular signal-regulated kinases (ERKs). 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 66-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 12145054-7 2002 These results indicate that midazolam induces the expression of c-Fos and EGR-1, by activation of ERKs through a mechanism independent from gamma-aminobutyric acid(A) receptors, in PC12 cells, and suggest the possibility that midazolam can induce long-term changes of neural functions by changing gene expression. Midazolam 28-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 12145054-7 2002 These results indicate that midazolam induces the expression of c-Fos and EGR-1, by activation of ERKs through a mechanism independent from gamma-aminobutyric acid(A) receptors, in PC12 cells, and suggest the possibility that midazolam can induce long-term changes of neural functions by changing gene expression. Midazolam 226-235 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 12148930-0 2002 Dissociation of conditioned locomotion and Fos induction in response to stimuli formerly paired with cocaine. Cocaine 101-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 12148930-4 2002 In contrast, pairing of cocaine with generalized contextual cues (whole room) produced conditioned locomotion and Fos activation in the prelimbic portion of prefrontal cortex and the nucleus accumbens core. Cocaine 24-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 12148940-0 2002 Conditioned Fos expression following morphine-paired contextual cue exposure is environment specific. Morphine 37-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-15 12507392-0 2002 Dexamethasone attenuates by colchicine induced Fos expression in the rat deep cerebellar and vestibular nuclei. Dexamethasone 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 12507392-0 2002 Dexamethasone attenuates by colchicine induced Fos expression in the rat deep cerebellar and vestibular nuclei. Colchicine 28-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 12507392-2 2002 The intent of the present study was to find out whether dexamethasone pretreatment may affect the induction of Fos protein in cell nuclei of the cerebellar vestibular neuronal complex (CVNC) elicited by central administration of colchicine. Dexamethasone 56-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 12507392-2 2002 The intent of the present study was to find out whether dexamethasone pretreatment may affect the induction of Fos protein in cell nuclei of the cerebellar vestibular neuronal complex (CVNC) elicited by central administration of colchicine. Colchicine 229-239 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 12507392-9 2002 However, in colchicine treated animals, which exhibited a large number of Fos-positive cells over the entire CVNC, the dexamethasone elicited a substantial reduction in the number of the Fos-immunoreactive cells over the CVNC. Colchicine 12-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 12507392-9 2002 However, in colchicine treated animals, which exhibited a large number of Fos-positive cells over the entire CVNC, the dexamethasone elicited a substantial reduction in the number of the Fos-immunoreactive cells over the CVNC. Colchicine 12-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 187-190 12507392-9 2002 However, in colchicine treated animals, which exhibited a large number of Fos-positive cells over the entire CVNC, the dexamethasone elicited a substantial reduction in the number of the Fos-immunoreactive cells over the CVNC. Dexamethasone 119-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 12507392-9 2002 However, in colchicine treated animals, which exhibited a large number of Fos-positive cells over the entire CVNC, the dexamethasone elicited a substantial reduction in the number of the Fos-immunoreactive cells over the CVNC. Dexamethasone 119-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 187-190 12507392-10 2002 Distinct dexamethasone dependent reduction (50-90%) of Fos-immunoreactivity was observed in each of the deep cerebellar nuclei. Dexamethasone 9-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 12507392-12 2002 From these, maximal Fos-inhibition by dexamethasone was recognized in the medial vestibular nucleus, however, even in this case the number of suppressed cells did not exceed 50%. Dexamethasone 38-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 12507392-14 2002 The results provide for the first time evidence about the dexamethasone dependent reduction of Fos-immunoreactivity in the cells of the CVNC in response to stimulation elicited by colchicine. Dexamethasone 58-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 12507392-14 2002 The results provide for the first time evidence about the dexamethasone dependent reduction of Fos-immunoreactivity in the cells of the CVNC in response to stimulation elicited by colchicine. Colchicine 180-190 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 12376248-3 2002 Kainic acid (0.01%) was stereotaxically injected into the hippocampus as a chemical stimulus, and immunohistochemistry method was used to show the expression of c-Fos in rat brain. Kainic Acid 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 12115667-2 2002 Hypotension-sensitive SPN, identified by immunoreactivity (IR) to the product of the immediate early gene c-fos and to choline acetyltransferase, were localized in the intermediolateral cell column of thoracic and upper lumbar cord, particularly middle to lower thoracic cord. spn 22-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 12270045-0 2002 Evidence that metyrapone can act as a stressor: effect on pituitary-adrenal hormones, plasma glucose and brain c-fos induction. Metyrapone 14-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 12135782-4 2002 The noxious thermal stimulus-evoked c-Fos expression level was reduced by APV and/or CNQX, while Zif/268 expression was hardly changed. apv 74-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 12131378-13 2002 Testosterone replacement significantly reduced the expression of c-fos mRNA in the medial preoptic area (p <0.05). Testosterone 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 12135782-4 2002 The noxious thermal stimulus-evoked c-Fos expression level was reduced by APV and/or CNQX, while Zif/268 expression was hardly changed. 6-Cyano-7-nitroquinoxaline-2,3-dione 85-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 12135782-5 2002 Both c-Fos and Zif/268 expressions following formalin injection were reduced by APV alone and APV+CNQX, but not by CNQX alone. Formaldehyde 45-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 5-10 12135782-5 2002 Both c-Fos and Zif/268 expressions following formalin injection were reduced by APV alone and APV+CNQX, but not by CNQX alone. 6-Cyano-7-nitroquinoxaline-2,3-dione 98-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 5-10 12135782-7 2002 These findings suggest that NMDA and AMPA/KA receptors are differentially involved in c-Fos and Zif/268 expression in the spinal dorsal horn following noxious thermal, formalin and mechanical stimulation. Formaldehyde 168-176 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 12174389-11 2002 The intensity of c-fos, c-jun and cyclin D(1) expression of HSCs treated with 10(-7)mol/L genistein for 48 h was also significantly decreased compared with the controls. Genistein 90-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 12174389-12 2002 CONCLUSION: Genistein influences proliferation of HSC, suppresses the expression of alpha-SMA in HSC and t inhibits the intensity of c-fos, c-jun and cyclin D(1) expression of HSCs. Genistein 12-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 12089747-0 2002 Effects of MK-801 and morphine on spinal C-Fos expression during the development of neuropathic pain. Dizocilpine Maleate 11-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 12093589-1 2002 We have characterized the effects of chronic clozapine and haloperidol treatments on the expression of fos (c-fos, fosB, fra-2) and jun (c-jun, junB, junD) family genes in the rat forebrain. Clozapine 45-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-106 12093589-1 2002 We have characterized the effects of chronic clozapine and haloperidol treatments on the expression of fos (c-fos, fosB, fra-2) and jun (c-jun, junB, junD) family genes in the rat forebrain. Clozapine 45-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 12093589-1 2002 We have characterized the effects of chronic clozapine and haloperidol treatments on the expression of fos (c-fos, fosB, fra-2) and jun (c-jun, junB, junD) family genes in the rat forebrain. Haloperidol 59-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-106 12093589-1 2002 We have characterized the effects of chronic clozapine and haloperidol treatments on the expression of fos (c-fos, fosB, fra-2) and jun (c-jun, junB, junD) family genes in the rat forebrain. Haloperidol 59-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 12093589-3 2002 Chronic clozapine treatment with a 6 d washout period induced the expression of several fos and jun family genes in cortical regions, including the prefrontal cortex (PFC), and in the caudate putamen and nucleus accumbens. Clozapine 8-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 12093589-5 2002 Chronic administration of haloperidol upregulated fos and jun family mRNA expression that was detectable 24 h and 6 d after cessation of the treatment mainly in the cortex. Haloperidol 26-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 12093589-7 2002 Thus, chronic treatments with clozapine and haloperidol induce a long-lasting enhancement of fos and jun family transcription factors that continues for several days after the cessation of the treatments in the cortex. Clozapine 30-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 12093589-7 2002 Thus, chronic treatments with clozapine and haloperidol induce a long-lasting enhancement of fos and jun family transcription factors that continues for several days after the cessation of the treatments in the cortex. Haloperidol 44-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 12106677-0 2002 Adrenal medullary transplants reduce formalin-evoked c-fos expression in the rat spinal cord. Formaldehyde 37-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 12106677-3 2002 In order to address this, expression of neural activity marker c-fos in response to intraplantar formalin was evaluated in animals with intrathecal adrenal medullary or control striated muscle transplants. Formaldehyde 97-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 12080023-6 2002 Administration of progesterone (P4; 16 mg/kg s.c./day) beginning on Day 20 (to maintain elevated plasma P4 levels) prevented the onset of labor and blocked the expected rise in c-fos, fosB, fra-1, fra-2, and junB expression on Day 23. Progesterone 18-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 177-182 12080023-7 2002 In contrast, administration of the progesterone receptor antagonist RU486 (10 mg/kg s.c.) on Day 19 induced preterm labor and a premature increase in mRNA levels of c-fos, fra-1, fra-2, and junB. Mifepristone 68-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 12128154-3 2002 In this study we investigated the effects of cocaine on c-fos and mu-opioid receptor mRNA levels in primary cortical astrocyte cultures, using RT-PCR and quantitative solution hybridization assays. Cocaine 45-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 12128154-6 2002 Our data suggest that cocaine differentially modulates c-fos and opioid signaling in astrocyte cell culture. Cocaine 22-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 12128156-1 2002 Psychostimulants such as cocaine have been shown to regulate c-fos and opioid gene expression in male rats. Cocaine 25-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 12128156-4 2002 The c-fos mRNA levels were increased in intact male and female rats after 30min or 3h of one single cocaine injection and after 14 days of single daily cocaine injections. Tritium 83-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 12128156-4 2002 The c-fos mRNA levels were increased in intact male and female rats after 30min or 3h of one single cocaine injection and after 14 days of single daily cocaine injections. Cocaine 100-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 12128156-4 2002 The c-fos mRNA levels were increased in intact male and female rats after 30min or 3h of one single cocaine injection and after 14 days of single daily cocaine injections. Cocaine 152-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 12086933-8 2002 Intracarotid glucose infusions stimulated c-fos expression in the same areas that expressed GK. Glucose 13-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 12072385-7 2002 Furthermore, induction of c-fos mRNA in limbic cortexes (but not hippocampus) was positively correlated with progesterone and negatively correlated with ACTH levels. Progesterone 109-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 12110967-3 2002 Microinjections (1 microl) of histamine, serotonin (5-HT), nicotine, capsaicin, or formalin each elicited similar distributions of Fos-like immunoreactivity (FLI) in laminae I-II of the ipsilateral superficial dorsal horn, with little or no FLI in deeper laminae or contralaterally. Histamine 30-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-134 12110967-3 2002 Microinjections (1 microl) of histamine, serotonin (5-HT), nicotine, capsaicin, or formalin each elicited similar distributions of Fos-like immunoreactivity (FLI) in laminae I-II of the ipsilateral superficial dorsal horn, with little or no FLI in deeper laminae or contralaterally. Nicotine 59-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-134 12110967-3 2002 Microinjections (1 microl) of histamine, serotonin (5-HT), nicotine, capsaicin, or formalin each elicited similar distributions of Fos-like immunoreactivity (FLI) in laminae I-II of the ipsilateral superficial dorsal horn, with little or no FLI in deeper laminae or contralaterally. Capsaicin 69-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-134 12110967-3 2002 Microinjections (1 microl) of histamine, serotonin (5-HT), nicotine, capsaicin, or formalin each elicited similar distributions of Fos-like immunoreactivity (FLI) in laminae I-II of the ipsilateral superficial dorsal horn, with little or no FLI in deeper laminae or contralaterally. Formaldehyde 83-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-134 23604908-8 2002 For CR"s action, we found that in the nucleus of aged rats, AP-1 activation was blunted by decreasing c-Jun and c-Fos levels and inhibiting c-Jun protein phosphorylation. Chromium 4-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 12184738-0 2002 7-Hydroxy-N,N"-di-n-propyl-2-aminotetraline, a preferential dopamine D3 agonist, induces c-fos mRNA expression in the rat cerebellum. 7-hydroxy-n,n"-di-n-propyl-2-aminotetraline 0-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 12184738-0 2002 7-Hydroxy-N,N"-di-n-propyl-2-aminotetraline, a preferential dopamine D3 agonist, induces c-fos mRNA expression in the rat cerebellum. Dopamine 60-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 12184738-1 2002 The effects of a preferential dopamine D3 receptor agonist 7-hydroxy-N,N"-di-n-propyl-2-aminotetralin (7-OH-DPAT) on c-fos mRNA expression in the rat cerebellum were studied by Northern blot analysis. 7-hydroxy-2-N,N-dipropylaminotetralin 59-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 12184738-1 2002 The effects of a preferential dopamine D3 receptor agonist 7-hydroxy-N,N"-di-n-propyl-2-aminotetralin (7-OH-DPAT) on c-fos mRNA expression in the rat cerebellum were studied by Northern blot analysis. 7-hydroxy-2-N,N-dipropylaminotetralin 103-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 12184738-2 2002 7-OH-DPAT (0.003-10 mg/kg, s.c.) markedly increased c-fos mRNA expression in the cerebellum, while its effects in the striatum, nucleus accumbens, and frontal cortex were negligible. 7-hydroxy-2-N,N-dipropylaminotetralin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 12184738-3 2002 The effect of 7-OH-DPAT on cerebellar c-fos mRNA expression was dose-dependent and statistically significant at doses of 0.3 mg/kg or more. 7-hydroxy-2-N,N-dipropylaminotetralin 14-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 12184738-6 2002 Furthermore, dopaminergic denervation by 6-hydroxydopamine did not inhibit but rather potentiated the 7-OH-DPAT-induced c-fos mRNA expression in the cerebellum. Oxidopamine 41-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 12184738-6 2002 Furthermore, dopaminergic denervation by 6-hydroxydopamine did not inhibit but rather potentiated the 7-OH-DPAT-induced c-fos mRNA expression in the cerebellum. 7-hydroxy-2-N,N-dipropylaminotetralin 102-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 12184738-7 2002 These findings suggest that 7-OH-DPAT increases c-fos mRNA expression in the rat cerebellum, probably through postsynaptic dopamine D3 receptor activation. 7-hydroxy-2-N,N-dipropylaminotetralin 28-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 12393948-0 2002 C-fos antisense oligodeoxynucleotide decreases subcutaneous bee venom injection-induced nociceptive behavior and fos expression in the rat. Oligodeoxyribonucleotides 16-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 12393948-0 2002 C-fos antisense oligodeoxynucleotide decreases subcutaneous bee venom injection-induced nociceptive behavior and fos expression in the rat. Oligodeoxyribonucleotides 16-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 2-5 12393948-1 2002 Oligodeoxynucleotide complementary to c-fos mRNA was applied to characterize its effect on the spinal cord Fos expression and relevant nociceptive behaviors challenged by subcutaneous injection of bee venom to the rat hind paw. Oligodeoxyribonucleotides 0-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 12393948-5 2002 At the same time, ASO treatment also significantly decreased the expression of Fos protein within the lumbar region of the spinal cord ipsilateral to the injection. 1,5-anhydroglucitol 18-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 12117572-4 2002 The food restriction regimen that augments drug reward also increases the induction of c-fos, by intracerebroventricular amphetamine, in limbic forebrain dopamine (DA) terminal areas. Amphetamine 121-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 12117572-4 2002 The food restriction regimen that augments drug reward also increases the induction of c-fos, by intracerebroventricular amphetamine, in limbic forebrain dopamine (DA) terminal areas. Dopamine 154-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 12117572-4 2002 The food restriction regimen that augments drug reward also increases the induction of c-fos, by intracerebroventricular amphetamine, in limbic forebrain dopamine (DA) terminal areas. Dopamine 164-166 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 12117580-6 2002 2-Deoxy-D-glucose (2DG) increases c-fos expression in orexin-A neurons in the DMN, and DMNL eliminated the orexigenic effect of 2DG. Deoxyglucose 0-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 12117580-6 2002 2-Deoxy-D-glucose (2DG) increases c-fos expression in orexin-A neurons in the DMN, and DMNL eliminated the orexigenic effect of 2DG. Deoxyglucose 19-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 12052441-0 2002 Sevoflurane suppresses noxious stimulus-evoked expression of Fos-like immunoreactivity in the rat spinal cord via activation of endogenous opioid systems. Sevoflurane 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 12052441-2 2002 Formalin injection into the hindpaw of the rat induces the nocifensive flinching behavior and the expression of Fos-like immunoreactivity (Fos-LI) in the spinal cord. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 12052441-2 2002 Formalin injection into the hindpaw of the rat induces the nocifensive flinching behavior and the expression of Fos-like immunoreactivity (Fos-LI) in the spinal cord. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 12052441-3 2002 Sevoflurane significantly suppressed the flinching behavior and decreased the number of Fos-LI neurons in the dorsal horn of spinal cord compared with the control group. Sevoflurane 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 12052441-4 2002 Moreover, pretreatment with intraperitoneal naloxone plus naltrexone antagonized the suppression of flinching behavior and the decrease of the number of Fos-LI neurons produced by 3% sevoflurane. Naloxone 44-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-156 12052441-4 2002 Moreover, pretreatment with intraperitoneal naloxone plus naltrexone antagonized the suppression of flinching behavior and the decrease of the number of Fos-LI neurons produced by 3% sevoflurane. Naltrexone 58-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-156 12052441-4 2002 Moreover, pretreatment with intraperitoneal naloxone plus naltrexone antagonized the suppression of flinching behavior and the decrease of the number of Fos-LI neurons produced by 3% sevoflurane. Sevoflurane 183-194 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-156 12010763-0 2002 Quinine and citric acid elicit distinctive Fos-like immunoreactivity in the rat nucleus of the solitary tract. Quinine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 12010763-0 2002 Quinine and citric acid elicit distinctive Fos-like immunoreactivity in the rat nucleus of the solitary tract. Citric Acid 12-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 12010763-1 2002 The present experiment investigated Fos-like immunoreactivity (FLI) in the nucleus of the solitary tract (NST) after intraoral infusions of 0.1 M citric acid, 0.3 M NaCl, and 0.3-30 mM quinine monohydrochloride (QHCl) in awake, behaving rats. Citric Acid 146-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-39 12105084-2 2002 Using in situ hybridization, the present report demonstrates that NO released by sodium nitroprusside regulates egr-1, c-fos, and junB immediate early gene expression in rat forebrain. Nitroprusside 81-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 12040067-5 2002 To identify the neuronal groups involved in the fever-inducing pathway, we first investigated Fos expression in medullary regions of rats after central administrations of PGE(2). Prostaglandins E 171-174 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 12040067-6 2002 PGE(2) application to the lateral ventricle or directly to the POA strikingly increased the number of Fos-positive neurons in the rostral part of the raphe pallidus nucleus (rRPa). Prostaglandins E 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 12093623-0 2002 CO(2)-induced c-Fos expression in brainstem preprotachykinin mRNA containing neurons. co(2) 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 12093623-3 2002 Experiments were performed in 21-day-old rats exposed to 12% CO(2) for 1 h. c-Fos expression was identified by IHH on free floating sections (40 microm) that were mounted and then hybridized with anti-sense 35S labeled ribonucleotide probe of the rat preprotachykinin A (PPT-A) gene. co(2) 61-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 12093623-3 2002 Experiments were performed in 21-day-old rats exposed to 12% CO(2) for 1 h. c-Fos expression was identified by IHH on free floating sections (40 microm) that were mounted and then hybridized with anti-sense 35S labeled ribonucleotide probe of the rat preprotachykinin A (PPT-A) gene. Sulfur-35 207-210 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 12093623-3 2002 Experiments were performed in 21-day-old rats exposed to 12% CO(2) for 1 h. c-Fos expression was identified by IHH on free floating sections (40 microm) that were mounted and then hybridized with anti-sense 35S labeled ribonucleotide probe of the rat preprotachykinin A (PPT-A) gene. 4,4',4''-(4-propyl-((1)H)-pyrazole-1,3,5-triyl) tris-phenol 271-274 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 12031688-7 2002 Finally, BVA treatment significantly suppressed adjuvant-induced Fos expression in the lumbar spinal cord at 3 weeks post-adjuvant injection. Trihydroxy[(N-Hydroxybenzamidato)oxo]vanadate 9-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 12475444-0 2002 Effects of selenium and iodine on the expression of c-fos and c-jun mRNA and their proteins in cultured rat hippocampus neurons. Selenium 11-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 12475444-0 2002 Effects of selenium and iodine on the expression of c-fos and c-jun mRNA and their proteins in cultured rat hippocampus neurons. Iodine 24-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 12475444-1 2002 OBJECTIVE: To study the effect of selenium (Se) and iodine (I) and the compound of both on the proto-oncogenes c-fos and c-jun mRNA and their protein expression in the cultured rat hippocampus neurons. Selenium 34-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 12475444-1 2002 OBJECTIVE: To study the effect of selenium (Se) and iodine (I) and the compound of both on the proto-oncogenes c-fos and c-jun mRNA and their protein expression in the cultured rat hippocampus neurons. Iodine 52-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 12475444-3 2002 The expression of the mRNA of c-fos, c-jun in hippocampus neurons cultured for 1, 3, 5, 7 and 10 d were studied using both in situ hybridization and SABC immunohistochemical technique. sabc 149-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 12089747-0 2002 Effects of MK-801 and morphine on spinal C-Fos expression during the development of neuropathic pain. Morphine 22-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 12019329-8 2002 Formalin injection induced c-Fos in approximately 80% of projection neurons with the NK1 receptor and in 25-45% of those without it. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 11992475-0 2002 Effects of somatosensory cortical stimulation on expression of c-Fos in rat medullary dorsal horn in response to formalin-induced noxious stimulation. Formaldehyde 113-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 11992475-1 2002 We examined the effects of epidural electrical stimulation of primary (SI) and secondary (SII) somatosensory cortex on expression of c-Fos protein in rat medullary dorsal horn neurons (Vc; trigeminal nucleus caudalis) in response to formalin-induced noxious stimulation. Formaldehyde 233-241 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 11992475-3 2002 SII stimulation at 1.0 mA immediately after injection of formalin, significantly decreased the number of Fos-positive cells in the right VcI/II by 32.4%. Formaldehyde 57-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-108 11992475-5 2002 SII stimulation at 0.5 and 1.0 mA 2 hr after injection of formalin, significantly decreased the number of Fos-positive cells in the right VcI/II by 47.9% and 40.8%, but significantly increased the number of Fos-positive cells in the right VcIII/IV by 178.8% and 324.3%, respectively. Formaldehyde 58-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 12020684-4 2002 After intracerebroventricular administration of 3-methoxytyramine, significantly more neurones expressed c-Fos in mesocortico-limbic dopamine areas including frontal cortex, medial prefrontal cortex, parietal cortex, piriform cortex, the nucleus accumbens shell, and ventral tegmental area. 3-methoxytyramine 48-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 11997149-0 2002 Contrasting effects of dopamine antagonists and frequency reduction on Fos expression induced by lateral hypothalamic stimulation. Dopamine 23-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 11997149-2 2002 Fos expression was also examined in stimulated and unstimulated rats pretreated with SCH 23390 (a dopamine D1 antagonist) or spiperone (a D2-like antagonist), at doses known to greatly inhibit responding for self-stimulation. SCH 23390 85-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 11997149-6 2002 Pretreatment with SCH 23390 (0.1 mg/kg) inhibited stimulation-induced Fos expression in some key dopamine terminal areas, such as the nucleus accumbens (core and shell) and medial caudate-putamen, but not in directly driven neurons near the stimulation site. SCH 23390 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 11997149-6 2002 Pretreatment with SCH 23390 (0.1 mg/kg) inhibited stimulation-induced Fos expression in some key dopamine terminal areas, such as the nucleus accumbens (core and shell) and medial caudate-putamen, but not in directly driven neurons near the stimulation site. Dopamine 97-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 12020684-4 2002 After intracerebroventricular administration of 3-methoxytyramine, significantly more neurones expressed c-Fos in mesocortico-limbic dopamine areas including frontal cortex, medial prefrontal cortex, parietal cortex, piriform cortex, the nucleus accumbens shell, and ventral tegmental area. Dopamine 133-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 12269402-3 2002 Additionally, repeated administrations of nicotine transiently induced the c-fos and c-jun mRNA levels after the first-third nicotine administration, and the c-fos and c-jun mRNA levels were returned to the basal level after the seventh administration of nicotine. Nicotine 42-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 12269402-3 2002 Additionally, repeated administrations of nicotine transiently induced the c-fos and c-jun mRNA levels after the first-third nicotine administration, and the c-fos and c-jun mRNA levels were returned to the basal level after the seventh administration of nicotine. Nicotine 42-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 12269402-3 2002 Additionally, repeated administrations of nicotine transiently induced the c-fos and c-jun mRNA levels after the first-third nicotine administration, and the c-fos and c-jun mRNA levels were returned to the basal level after the seventh administration of nicotine. Nicotine 125-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 12269402-3 2002 Additionally, repeated administrations of nicotine transiently induced the c-fos and c-jun mRNA levels after the first-third nicotine administration, and the c-fos and c-jun mRNA levels were returned to the basal level after the seventh administration of nicotine. Nicotine 125-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 12269402-6 2002 Immunohistochemical analysis showed that the increase of c-Fos and c-Jun proteins by repeated nicotine administrations is mostly medulla specific, while Fra-2 immuno reactivity was shown both in medulla and cortex. Nicotine 94-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 12269402-9 2002 These results suggest that proENK mRNA expression induced by repeated nicotine administrations may be mediated by AP-1 proteins, such as c-Fos, c-Jun and Fra-2 rather than CREB via interacting to the ENKCRE-2 DNA binding domain in rat adrenal medulla. Nicotine 70-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 12006386-10 2002 Induction of c-fos expression and enhanced AP-1 binding activity by lysoPC were also inhibited by DPI and NAC. diphenyleneiodonium 98-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 12006386-10 2002 Induction of c-fos expression and enhanced AP-1 binding activity by lysoPC were also inhibited by DPI and NAC. Acetylcysteine 106-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 11976269-3 2002 Here we have studied the expression of Fos after administration of morphine and during morphine withdrawal in the rat hypothalamic PVN and SON. Morphine 67-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 11976269-7 2002 Acute morphine administration produced an increase in Fos expression at hypothalamic nuclei and in the brainstem regions, and tolerance developed towards this effect. Morphine 6-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 11976269-8 2002 Precipitated morphine withdrawal induced marked Fos immunoreactivity within the PVN and SON. Morphine 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 11976269-10 2002 Moreover, catecholaminergic-positive neurons in the brainstem showed a significant increase in Fos expression in response to morphine withdrawal. Morphine 125-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 12009778-0 2002 Amphetamine-induced Fos expression is evident in gamma-aminobutyric acid neurons in the globus pallidus and entopeduncular nucleus in rats treated with intrastriatal c-fos antisense oligodeoxynucleotides. gamma-Aminobutyric Acid 49-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 12039681-3 2002 Ace and Meth, but not IL-1, inhibited AChE activity, while all three compounds; (1) increased ACh and CRF mRNA levels in and CRF release from; (2) activated the CRE promoter region of CRF-gene in: and (3) increased cFos binding to the AP-1 region of the CRF-gene in the hypothalamus. Methamphetamine 8-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 215-219 12009778-0 2002 Amphetamine-induced Fos expression is evident in gamma-aminobutyric acid neurons in the globus pallidus and entopeduncular nucleus in rats treated with intrastriatal c-fos antisense oligodeoxynucleotides. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 12009778-0 2002 Amphetamine-induced Fos expression is evident in gamma-aminobutyric acid neurons in the globus pallidus and entopeduncular nucleus in rats treated with intrastriatal c-fos antisense oligodeoxynucleotides. gamma-Aminobutyric Acid 49-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-171 12009778-0 2002 Amphetamine-induced Fos expression is evident in gamma-aminobutyric acid neurons in the globus pallidus and entopeduncular nucleus in rats treated with intrastriatal c-fos antisense oligodeoxynucleotides. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-171 12009778-0 2002 Amphetamine-induced Fos expression is evident in gamma-aminobutyric acid neurons in the globus pallidus and entopeduncular nucleus in rats treated with intrastriatal c-fos antisense oligodeoxynucleotides. Oligodeoxyribonucleotides 182-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 12009778-0 2002 Amphetamine-induced Fos expression is evident in gamma-aminobutyric acid neurons in the globus pallidus and entopeduncular nucleus in rats treated with intrastriatal c-fos antisense oligodeoxynucleotides. Oligodeoxyribonucleotides 182-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-171 12009778-2 2002 Striatal efferent activity was suppressed by intrastriatal infusions of antisense oligodeoxynucleotide targeted to the messenger RNA of the immediate early gene, c-fos. Oligodeoxyribonucleotides 82-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 12009778-3 2002 This suppression produced robust rotational behavior and expression of Fos in the ipsilateral GP and EP following amphetamine challenge. Amphetamine 114-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 12009778-4 2002 The expression of Fos in the ipsilateral GP and EP following amphetamine challenge is not observed in naive or control antisense-treated animals. Amphetamine 61-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 12009778-5 2002 Quantitative analysis revealed that a majority of the amphetamine-activated (Fos-immunoreactive) neurons in the GP and EP express GABA. Amphetamine 54-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-80 12009778-5 2002 Quantitative analysis revealed that a majority of the amphetamine-activated (Fos-immunoreactive) neurons in the GP and EP express GABA. gamma-Aminobutyric Acid 130-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-80 12059975-9 2002 Moreover, cells expressing c-fos immunoreactivity were significantly more numerous in ciproxifan- or thioperamide-treated rats than in controls, although no colocalization of anti-c-fos and anti-ChAT immunoreactivity was observed. ciproxifan 86-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 12138252-2 2002 After topical application of capsaicin (1%) to the sciatic nerve, FOS-like immunoreactive (FOS-LI) neurons were observed, chiefly in the superficial laminae of the lumbar dorsal horn. Capsaicin 29-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 12059975-9 2002 Moreover, cells expressing c-fos immunoreactivity were significantly more numerous in ciproxifan- or thioperamide-treated rats than in controls, although no colocalization of anti-c-fos and anti-ChAT immunoreactivity was observed. thioperamide 101-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 11961044-6 2002 In addition, increased RhoA protein, Rho-kinase, and c-fos gene expression, and myosin light chain phosphorylation were suppressed by Y-27632 and candesartan. Y 27632 134-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 11961044-6 2002 In addition, increased RhoA protein, Rho-kinase, and c-fos gene expression, and myosin light chain phosphorylation were suppressed by Y-27632 and candesartan. candesartan 146-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 12133320-0 2002 [The effect of triptolide on the proliferation of airway smooth muscle and the expression of c-fos and c-jun in asthmatic rats]. triptolide 15-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 12133320-12 2002 The remarkable inhibition of ASM proliferation by triptolide was probably due to its inhibitory effect on the expression of c-fos and c-jun. triptolide 50-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 12008016-5 2002 CPA pretreatment significantly attenuated the PCP-induced increase in c-fos mRNA levels in the medial prefrontal cortex and nucleus accumbens. N(6)-cyclopentyladenosine 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 12008016-5 2002 CPA pretreatment significantly attenuated the PCP-induced increase in c-fos mRNA levels in the medial prefrontal cortex and nucleus accumbens. Phencyclidine 46-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 11931859-0 2002 Heroin sensitization as mapped by c-Fos immunoreactivity in the rat striatum. Heroin 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 11931859-2 2002 Acute injection of heroin to drug-naive rats caused significant induction of c-Fos protein in the nucleus accumbens shell, whereas the same dose of heroin given to drug-sensitized rats significantly increased c-Fos immunoreactivity in the dorsomedial caudate-putamen. Heroin 19-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 11931859-2 2002 Acute injection of heroin to drug-naive rats caused significant induction of c-Fos protein in the nucleus accumbens shell, whereas the same dose of heroin given to drug-sensitized rats significantly increased c-Fos immunoreactivity in the dorsomedial caudate-putamen. Heroin 19-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 209-214 11931859-3 2002 These results show that the heroin-induced pattern of c-Fos protein in the rat striatum differs according to the rat"s drug history. Heroin 28-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 12007921-1 2002 We have evaluated the effects of nefopam on the spinal c-Fos protein expression in the model of acute (noxious heat) and persistent (intraplantar injection of formalin) nociception in the rat. Formaldehyde 159-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 12007921-3 2002 formalin injection, c-Fos immunoreactive (c-Fos-IR) nuclei were preferentially located in the superficial (I-II) and deep (V-VI) laminae of the spinal dorsal horn of segments L4-L5, i.e. spinal areas containing numerous neurons responding exclusively, or not, to peripheral nociceptive stimuli. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 12007921-4 2002 The doses of 15 and 30 mg/kg (s.c.) of nefopam had significant reducing effects on the formalin-evoked spinal c-Fos protein expression (36+/-14% and 47+/-9% reduction of the total number of c-Fos-IR nuclei per section, respectively, P<0.05 for both). Formaldehyde 87-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 12007921-4 2002 The doses of 15 and 30 mg/kg (s.c.) of nefopam had significant reducing effects on the formalin-evoked spinal c-Fos protein expression (36+/-14% and 47+/-9% reduction of the total number of c-Fos-IR nuclei per section, respectively, P<0.05 for both). Formaldehyde 87-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 190-195 12007921-8 2002 Nefopam (15 mg/kg s.c.) significantly reduced the noxious heat-evoked spinal c-Fos protein expression (33+/-3% reduction of the total number of c-Fos-IR nuclei, P<0.0001). Nefopam 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 12007921-8 2002 Nefopam (15 mg/kg s.c.) significantly reduced the noxious heat-evoked spinal c-Fos protein expression (33+/-3% reduction of the total number of c-Fos-IR nuclei, P<0.0001). Nefopam 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-149 11999902-0 2002 Effects of MK-801 and electroconvulsive shock on c-Fos expression in the rat hippocampus and frontal cortex. Dizocilpine Maleate 11-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 12063085-3 2002 The number of c-Fos immunoreactive nuclei was evaluated in the lumbar spinal cord 90 min after carrageenin. Carrageenan 95-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 12063085-8 2002 In chronically treated RB101(S) rats, both acute RB101(S) and morphine reduced the total number of carrageenin-evoked c-Fos-immunoreactive nuclei. Morphine 62-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 12063085-8 2002 In chronically treated RB101(S) rats, both acute RB101(S) and morphine reduced the total number of carrageenin-evoked c-Fos-immunoreactive nuclei. Carrageenan 99-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 12063085-10 2002 reduced the total number of carrageenin-evoked c-Fos-immunoreactive nuclei with similar magnitude in naive and in morphine-tolerant (100 mg/kg/day for 3 days, s.c.) rats. Carrageenan 28-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 11858942-4 2002 In contrast, DMSO reduced the expression of immediate early genes (IEG) expression (c-myc, c-jun, c-fos, junB, egr-1) and inhibited mitogen-activated protein kinase (MEK) kinase/extracellular signal-regulated kinases (ERKs) and p38 phosphorylation. Dimethyl Sulfoxide 13-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 11922660-0 2002 Intrapericardial procaine affects volume expansion-induced fos immunoreactivity in unanesthetized rats. Procaine 17-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 11922660-9 2002 saline produced a significant increase in Fos expression in the nucleus of the solitary tract, the ventrolateral medulla, the area postrema, the locus coeruleus, the paraventricular nucleus of the hypothalamus, the perinuclear zone of the supraoptic nucleus, and oxytocin neurons in the supraoptic nucleus. Sodium Chloride 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 11922660-11 2002 procaine significantly blocked Fos expression produced by the volume expansion in the all of the regions examined except for the area postrema and the SON oxytocin neurons. Procaine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 11999902-1 2002 Both the N-methyl-D-aspartate (NMDA) receptor antagonist MK-801 and electroconvulsive shock (ECS) have been reported to induce c-Fos in rat brain. Dizocilpine Maleate 57-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 11999902-3 2002 To understand the mode of action of these treatments, the authors examined the effect of MK-801 and the interaction between MK-801 and ECS on the induction of c-Fos in the rat hippocampus and frontal cortex. Dizocilpine Maleate 124-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-164 11999902-4 2002 MK-801 induced c-Fos in these brain regions in a nonlinear dose-response relationship. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 11999902-7 2002 Pretreatment with MK-801 dose-dependently attenuated both the seizures and c-Fos expression by ECS. Dizocilpine Maleate 18-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 11999902-8 2002 However, at an MK-801 pretreatment dose of 8 mg/kg, which completely blocked ECS-induced seizure, the induction of c-Fos was not completely blocked, suggesting non-NMDA mediated pathways of the induction of c-Fos by ECS. Dizocilpine Maleate 15-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 207-212 11897101-9 2002 Thus, decreased water intake by VGX rats was accompanied by decreased Fos-ir in the parvocellular PVN after the same treatments, indicating a role for the abdominal vagus in thirst in response to signaling from gut osmoreceptors. Water 16-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 12539272-0 2002 [C-Fos expression in the hippocampus of rats with pentylenetetrazol-induced epilepsy] OBJECTIVE: To investigate the expression of c-FOS oncogene in rats hippocampus with pentylenetetrazol (PTZ)-induced generalized seizure. Pentylenetetrazole 50-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 1-6 12539272-0 2002 [C-Fos expression in the hippocampus of rats with pentylenetetrazol-induced epilepsy] OBJECTIVE: To investigate the expression of c-FOS oncogene in rats hippocampus with pentylenetetrazol (PTZ)-induced generalized seizure. Pentylenetetrazole 50-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 12539272-0 2002 [C-Fos expression in the hippocampus of rats with pentylenetetrazol-induced epilepsy] OBJECTIVE: To investigate the expression of c-FOS oncogene in rats hippocampus with pentylenetetrazol (PTZ)-induced generalized seizure. Pentylenetetrazole 170-187 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 1-6 12539272-0 2002 [C-Fos expression in the hippocampus of rats with pentylenetetrazol-induced epilepsy] OBJECTIVE: To investigate the expression of c-FOS oncogene in rats hippocampus with pentylenetetrazol (PTZ)-induced generalized seizure. Pentylenetetrazole 170-187 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 12539272-0 2002 [C-Fos expression in the hippocampus of rats with pentylenetetrazol-induced epilepsy] OBJECTIVE: To investigate the expression of c-FOS oncogene in rats hippocampus with pentylenetetrazol (PTZ)-induced generalized seizure. Pentylenetetrazole 189-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 1-6 12539272-5 2002 The c-FOS protein expression was observed at 2 h or 4 h using peroxidase-labelled streptavidin biotin (LSAB) staining techniques. Biotin 95-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 12539272-9 2002 There was low level c-FOS protein expression in hippocampal pyramidal neurons and granule cells of the dentate gyrus.C-FOS expression was upregulated at the 2nd and 4th hour post-PTZ injection. Pentylenetetrazole 179-182 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 12539272-9 2002 There was low level c-FOS protein expression in hippocampal pyramidal neurons and granule cells of the dentate gyrus.C-FOS expression was upregulated at the 2nd and 4th hour post-PTZ injection. Pentylenetetrazole 179-182 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 12539272-10 2002 Nimodipine could reduce c-FOS protein expression induced by the PTZ-induced generalized seizure at 2nd hour post-PTZ injection (P<0.01). Nimodipine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 12539272-10 2002 Nimodipine could reduce c-FOS protein expression induced by the PTZ-induced generalized seizure at 2nd hour post-PTZ injection (P<0.01). Pentylenetetrazole 64-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 12539272-10 2002 Nimodipine could reduce c-FOS protein expression induced by the PTZ-induced generalized seizure at 2nd hour post-PTZ injection (P<0.01). Pentylenetetrazole 113-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 11897103-0 2002 2,3,7,8-Tetrachlorodibenzo-p-dioxin treatment induces c-Fos expression in the forebrain of the Long-Evans rat. Polychlorinated Dibenzodioxins 0-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 11897103-2 2002 In the present study, we examined c-Fos expression in the central nervous system (CNS) after administration of a lethal dose of TCDD to the adult Long-Evans rat to clarify if the CNS participates in TCDD-induced intoxication. Polychlorinated Dibenzodioxins 128-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 11879792-1 2002 The ability of amphetamine or cocaine to induce the expression of c-fos mRNA in a number of brain regions is greatly enhanced when these drugs are administered in a distinct and relatively novel environment, relative to when they are given in the home cage. Amphetamine 15-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 11879792-1 2002 The ability of amphetamine or cocaine to induce the expression of c-fos mRNA in a number of brain regions is greatly enhanced when these drugs are administered in a distinct and relatively novel environment, relative to when they are given in the home cage. Cocaine 30-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 11927369-3 2002 We employed in situ hybridization to explore the differences of c-fos mRNA activation in juvenile rats that had been allowed R&T play for a total of 30 min before sacrifice contrasted to animals with comparable histories that had received no play. O-beta-ribosyl(1''--2')adenosine-5''-phosphate 125-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 11840475-10 2002 The RVLM of six unanesthetized rats subjected to 2 hours of hydralazine-induced hypotension contained tenfold more c-Fos-ir DNPI/VGLUT2 neurons than that of six saline-treated controls. Hydralazine 60-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 11906706-7 2002 Western blot analysis showed that the levels of c-Fos and c-Jun protein expression, which increased at 1 h after partial hepatectomy, were also reduced by methotrexate. Methotrexate 155-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 11751895-8 2002 The mRNA levels of c-jun and c-fos in response to MIF were also inhibited by PD98059. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 77-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 11861123-7 2002 Cells in the molecular layer express Fos-like immunoreactivity following harmaline stimulation in a time and lobule specific manner; they do not appear to be activated in the electrical stimulation paradigm. Harmaline 73-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 12041912-9 2002 Northern and Western analyses indicated that 10(-6) M dexamethasone markedly increased the abundance of c-fos mRNA at 20 min and c-fos protein concentration at 60 min in 1,25(OH)2D3-treated UMR-106 cells but only slightly induced the abundance of c-jun mRNA. Dexamethasone 54-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 12041912-9 2002 Northern and Western analyses indicated that 10(-6) M dexamethasone markedly increased the abundance of c-fos mRNA at 20 min and c-fos protein concentration at 60 min in 1,25(OH)2D3-treated UMR-106 cells but only slightly induced the abundance of c-jun mRNA. Dexamethasone 54-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 12041912-10 2002 The addition of phorbol 12-myristate 13-acetate increased mRNA expression for both c-fos and 24-hydroxylase in 1,25(OH)2D3-treated UMR-106 cells. Tetradecanoylphorbol Acetate 16-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 11861123-8 2002 Following harmaline injections, there is an initial loss of Fos-like immunoreactivity in the cytoplasm of Purkinje cells; 90 min later, nuclear staining is observed in a few scattered Purkinje cells. Harmaline 10-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 11932076-0 2002 Vagotomy prevents morphine-induced reduction in Fos-like immunoreactivity in trigeminal spinal nucleus produced after TMJ injury in a sex-dependent manner. Morphine 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 11999722-6 2002 In the diagonal band of Broca and the perinuclear zone, the number of Fos-positive neurones was significantly increased after phenylephrine infusion. Phenylephrine 126-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 11932076-2 2002 Pretreatment with morphine greatly reduces Fos-LI produced in the dorsal paratrigeminal area (dPa5), ventrolateral pole of the subnucleus interpolaris/caudalis (Vi/Vc-vl) transition region, and laminae I-II at the subnucleus caudalis/upper cervical cord junction (Vc/C2) suggesting a role for these areas in processing pain signals from the TMJ region. Morphine 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 11932076-5 2002 By contrast, morphine-induced reduction in Fos-LI produced at the Vi/Vc-vl transition region was prevented by prior VgX in males and diestrus females, but not in proestrus females. Morphine 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 11932076-6 2002 Morphine inhibition of Fos-LI produced in laminae I-II at the Vc/C2 junction region was diminished in vagotomized males compared to intact animals, but not affected in females. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 11932076-7 2002 In an autonomic control area, the caudal ventrolateral medulla (CVLM), VgX reversed the morphine-induced reduction in Fos-LI in males and females similarly compared to their respective intact controls. Morphine 88-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 12039421-0 2002 Fos and nitric oxide synthase in rat brain with chronic mesostriatal dopamine deficiency: effects of nitroglycerin and hypoxia. Nitroglycerin 101-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 12039421-2 2002 Systemic administration of nitroglycerin (NTG) or mild hypoxia resulted in a decreased of c-fos expression in the dorsolateral part of the denervated neostriatum. Nitroglycerin 27-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 12039421-2 2002 Systemic administration of nitroglycerin (NTG) or mild hypoxia resulted in a decreased of c-fos expression in the dorsolateral part of the denervated neostriatum. Nitroglycerin 42-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 12039421-3 2002 However, in other brain structures NTG or mild hypoxia evoked sustained c-fos expression in NOS-containing neurons and in the sources catecholaminergic projections involved in the control of cardiovascular function. Nitroglycerin 35-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 11830185-3 2002 Immunocytochemical and in situ hybridization studies indicate that acute high-dose ethanol administration increases c-fos expression in GABAergic neurons within the CeA of the rat, suggesting activation of these neurons by ethanol. Ethanol 83-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 11830185-3 2002 Immunocytochemical and in situ hybridization studies indicate that acute high-dose ethanol administration increases c-fos expression in GABAergic neurons within the CeA of the rat, suggesting activation of these neurons by ethanol. Ethanol 223-230 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 11897260-2 2002 We previously found that gonadectomized male and female rats treated in adulthood with testosterone propionate (TP) showed equivalent Fos responses in the AOS to odors derived from estrous females. Testosterone Propionate 87-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-137 11850142-0 2002 Male rats exposed to cocaine in utero demonstrate elevated expression of Fos in the prefrontal cortex in response to environment. Cocaine 21-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-76 11850142-4 2002 In this report, we examined the effects of intravenous prenatal cocaine exposure on the expression of the immediate-early gene, c-fos, in the adolescent offspring to determine potential sites of disruption. Cocaine 64-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-133 11850142-5 2002 The expression of Fos protein was similar in unhandled rats prenatally treated with saline or cocaine. Sodium Chloride 84-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 11850142-5 2002 The expression of Fos protein was similar in unhandled rats prenatally treated with saline or cocaine. Cocaine 94-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 11850142-6 2002 Prenatal cocaine exposed rats that were handled, but not footshocked, however, demonstrated a dramatic selective increase in Fos expression in the ventral and medial prefrontal cortex. Cocaine 9-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-128 11850142-7 2002 A footshock-induced increase in Fos expression in the prefrontal cortex was noted in prenatal saline, but not prenatal cocaine rats. Sodium Chloride 94-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 11850142-7 2002 A footshock-induced increase in Fos expression in the prefrontal cortex was noted in prenatal saline, but not prenatal cocaine rats. Cocaine 119-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 11897260-2 2002 We previously found that gonadectomized male and female rats treated in adulthood with testosterone propionate (TP) showed equivalent Fos responses in the AOS to odors derived from estrous females. Testosterone Propionate 112-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-137 11850145-8 2002 Acute morphine withdrawal increased c-fos mRNA expression in the brain and the spinal cord, which was attenuated by pre-treatment of LY235959. Morphine 6-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 11882344-10 2002 There were increased numbers of Fos-immunoreactive cells in fetuses exposed to both ethanol and cocaine compared to cocaine binge only. Ethanol 84-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 11850145-8 2002 Acute morphine withdrawal increased c-fos mRNA expression in the brain and the spinal cord, which was attenuated by pre-treatment of LY235959. LY 235959 133-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 11869806-2 2002 We examined the time course of the effects of all-trans retinoic acid (RA) on HMOR and c-fos mRNA levels as determined by solution hybridization (using HMOR and rat c-fos riboprobes) in RNA extracts from SH-SY5Y cells. Tretinoin 71-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 11882344-10 2002 There were increased numbers of Fos-immunoreactive cells in fetuses exposed to both ethanol and cocaine compared to cocaine binge only. Cocaine 96-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 11882344-10 2002 There were increased numbers of Fos-immunoreactive cells in fetuses exposed to both ethanol and cocaine compared to cocaine binge only. Cocaine 116-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 11930235-0 2002 17beta-Estradiol inhibits vascular smooth muscle cell proliferation and c -fos expression: role of nitric oxide. Estradiol 0-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-78 11911955-2 2002 Administration of PGF2alpha increased c-Fos mRNA with a corresponding reduction in StAR mRNA. Dinoprost 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 11911955-4 2002 Co-transfection of c-Fos with the p-1862 StAR promoter caused a reduction in luciferase activity in the presence or absence of cAMP. Cyclic AMP 127-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 11788392-8 2002 PYK2 downregulation with antisense oligonucleotides blocked ANG II-induced c-Fos expression. Oligonucleotides 35-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 11792666-3 2002 Expression of Fos (as assessed by immunohistochemistry) was increased in the organum vasculosum of the lamina terminalis (OVLT), median preoptic nucleus (MnPO), subfornical organ (SFO), and supraoptic nucleus (SON) after water deprivation. Water 221-226 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 11792666-4 2002 After rehydration with water but before consumption of 1.8% NaCl, Fos expression in the SON disappeared and was partially reduced in the OVLT and MnPO. Water 23-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 11792666-4 2002 After rehydration with water but before consumption of 1.8% NaCl, Fos expression in the SON disappeared and was partially reduced in the OVLT and MnPO. Sodium Chloride 60-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 11792666-8 2002 The changes in Fos expression and PRA are consistent with a proposed role for ANG II in the control of the sodium appetite produced by water deprivation followed by rehydration with only water. Sodium 107-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-18 11792666-8 2002 The changes in Fos expression and PRA are consistent with a proposed role for ANG II in the control of the sodium appetite produced by water deprivation followed by rehydration with only water. Water 135-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-18 11812713-0 2002 Peripheral amitriptyline suppresses formalin-induced Fos expression in the rat spinal cord. Amitriptyline 11-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 11812713-0 2002 Peripheral amitriptyline suppresses formalin-induced Fos expression in the rat spinal cord. Formaldehyde 36-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 11812713-1 2002 UNLABELLED: We examined the effects of systemically, spinally, and peripherally administered amitriptyline on formalin-induced Fos immunoreactivity in the lumbar spinal cord. Amitriptyline 93-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-130 11812713-1 2002 UNLABELLED: We examined the effects of systemically, spinally, and peripherally administered amitriptyline on formalin-induced Fos immunoreactivity in the lumbar spinal cord. Formaldehyde 110-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-130 11812713-2 2002 Formalin (2.5%), injected subcutaneously into the rat hindpaw, increased Fos immunoreactivity in laminae I-II, III-IV, and V-VI of the dorsal L5 spinal cord. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-76 11812713-4 2002 Amitriptyline coadministered with the formalin reduced both flinching and biting/licking behaviors, and significantly reduced Fos immunoreactivity, particularly in laminae I-II. Amitriptyline 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-129 11812713-4 2002 Amitriptyline coadministered with the formalin reduced both flinching and biting/licking behaviors, and significantly reduced Fos immunoreactivity, particularly in laminae I-II. Formaldehyde 38-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-129 11812713-8 2002 Fos protein, an indicator of neuronal activity after noxious stimulation, is upregulated after formalin injection. Formaldehyde 95-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 11796524-1 2002 We have previously shown that PTH induction of c-fos expression in the rat osteoblastic cell line UMR 106-01 requires the phosphorylation of cAMP response element-binding protein (CREB) at serine 133. Serine 189-195 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 11886451-9 2002 However, pretreatment with the atypical APD clozapine did recapitulate the characteristic compartmental Fos pattern seen in response to typical APDs. Clozapine 44-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 11796656-2 2002 We tested the overall hypothesis that circulating gonadal steroids determine the gender differences in morphine- and MK-801-induced behavior and c-Fos expression. Steroids 58-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-150 11796656-3 2002 Morphine caused a greater expression of c-Fos in the striatum of intact males than of that females, which was independent of sex steroids. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 11796656-4 2002 MK-801 completely inhibited morphine-induced c-Fos in intact females but only caused partial inhibition in intact males; castrated males showed complete inhibition, which was reversed by testosterone, but gonadal steroids had no effect on this response in females. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 11796656-4 2002 MK-801 completely inhibited morphine-induced c-Fos in intact females but only caused partial inhibition in intact males; castrated males showed complete inhibition, which was reversed by testosterone, but gonadal steroids had no effect on this response in females. Morphine 28-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 11851354-7 2002 Furthermore, activation of ERK and Shc, and c- fos gene expression were significantly inhibited by AG1478 but not by cytochalasin D or PP1. RTKI cpd 99-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-50 11790521-6 2002 Inhibition of Fos expression in this region of the hippocampus, but not the cingulate and motor cortex, by means of antisense oligonucleotide treatment resulted in an impairment of spatial memory formation. Oligonucleotides 126-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 11839422-10 2002 Furthermore, an extensively greater number of Fos protein-LI cells were expressed both in superficial and deep laminae of the bilateral Vc and C1 of the spinal cord after subcutaneous injection of mustard oil into the whisker pad. mustard oil 197-208 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 11839422-11 2002 Fos protein expression after mustard oil injection was much stronger than that observed after any mechanical stimulation in the rats with IAN transection. mustard oil 29-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 11846624-8 2002 After treatment with Sch 58261, a considerable reduction in c-fos expression was observed in the ischemic hemispheres, whereas a limited effect was detected in the others. 5-amino-7-(2-phenylethyl)-2-(2-furyl)pyrazolo(4,3-e)-1,2,4-triazolo(1,5-c)pyrimidine 21-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 11754491-10 2002 Taken together with the demonstration that acute injection of this dose of mAMPH induces the immediate early gene zif/268 and Fos protein in barrel cortex, these data suggest that the prolonged behavioral activity involving the vibrissae contributes to the mAMPH-induced damage to S1 neurons. Methamphetamine 75-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-129 12561560-0 2002 [Effect of methylmercuric chloride on apoptosis and c-fos expression of brain glial cells]. methylmercuric chloride 11-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 12561560-3 2002 The expressed c-Fos protein began to increase in the nuclei of cultured glial cells at 0.5 h after MMC was added to DMEM medium for 10 min and increased markedly with time prolonged. Mitomycin 99-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 12561560-3 2002 The expressed c-Fos protein began to increase in the nuclei of cultured glial cells at 0.5 h after MMC was added to DMEM medium for 10 min and increased markedly with time prolonged. dmem medium 116-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 12561560-4 2002 The percentage of Fos-positive nuclei reached a peak at 2 h and declined gradually from 4 to 6 h. The percentage of Fos-positive glial nuclei in the group exposed to 0.32 mmol/L MMC was significantly higher than those in other groups, which were exposed to 0.00125, 0.005 and 0.02 mmol/L MMC respectively (P < 0.01) at 2 h after MMC was added. Mitomycin 178-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 12561560-4 2002 The percentage of Fos-positive nuclei reached a peak at 2 h and declined gradually from 4 to 6 h. The percentage of Fos-positive glial nuclei in the group exposed to 0.32 mmol/L MMC was significantly higher than those in other groups, which were exposed to 0.00125, 0.005 and 0.02 mmol/L MMC respectively (P < 0.01) at 2 h after MMC was added. Mitomycin 178-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 12561560-4 2002 The percentage of Fos-positive nuclei reached a peak at 2 h and declined gradually from 4 to 6 h. The percentage of Fos-positive glial nuclei in the group exposed to 0.32 mmol/L MMC was significantly higher than those in other groups, which were exposed to 0.00125, 0.005 and 0.02 mmol/L MMC respectively (P < 0.01) at 2 h after MMC was added. Mitomycin 288-291 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 12561560-4 2002 The percentage of Fos-positive nuclei reached a peak at 2 h and declined gradually from 4 to 6 h. The percentage of Fos-positive glial nuclei in the group exposed to 0.32 mmol/L MMC was significantly higher than those in other groups, which were exposed to 0.00125, 0.005 and 0.02 mmol/L MMC respectively (P < 0.01) at 2 h after MMC was added. Mitomycin 288-291 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 12561560-4 2002 The percentage of Fos-positive nuclei reached a peak at 2 h and declined gradually from 4 to 6 h. The percentage of Fos-positive glial nuclei in the group exposed to 0.32 mmol/L MMC was significantly higher than those in other groups, which were exposed to 0.00125, 0.005 and 0.02 mmol/L MMC respectively (P < 0.01) at 2 h after MMC was added. Mitomycin 288-291 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 12561560-4 2002 The percentage of Fos-positive nuclei reached a peak at 2 h and declined gradually from 4 to 6 h. The percentage of Fos-positive glial nuclei in the group exposed to 0.32 mmol/L MMC was significantly higher than those in other groups, which were exposed to 0.00125, 0.005 and 0.02 mmol/L MMC respectively (P < 0.01) at 2 h after MMC was added. Mitomycin 288-291 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 12561560-5 2002 It is concluded that MMC can induce the expression of c-fos and the apoptosis of glial cells in vitro. Mitomycin 21-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 21179853-1 2002 AIM AND METHODS: The method of labeled streptavidin biotin was used to study the expression of c-fos in various functional state of rat ovaries and its relationship with the levels of serum estradiol and progesterone. Biotin 52-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 21179853-1 2002 AIM AND METHODS: The method of labeled streptavidin biotin was used to study the expression of c-fos in various functional state of rat ovaries and its relationship with the levels of serum estradiol and progesterone. Estradiol 190-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 21179853-1 2002 AIM AND METHODS: The method of labeled streptavidin biotin was used to study the expression of c-fos in various functional state of rat ovaries and its relationship with the levels of serum estradiol and progesterone. Progesterone 204-216 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 11750902-6 2002 Pharmacological blockade of mGluRs with bilateral injections of a non-selective mGluR antagonist, (S)-alpha-methyl-4-carboxyphenylglycine (MCPG), into the dorsal striatum at 10 but not 0.4 nmol significantly attenuated amphetamine-stimulated c-fos mRNA expression in this area. (S)-MCPG 98-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 242-247 11750902-11 2002 Activation of the MCPG-sensitive mGluRs is required for the upregulation of transcription factor c-fos, although not zif/268, mRNA expression in the striatum in response to acute injection of amphetamine. Amphetamine 192-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 12553707-0 2002 Localization of c-Fos protein in the rat spinal cord after carrageenan treatment. Carrageenan 59-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 12553707-1 2002 We have characterized segmental and laminar distribution patterns of Fos-immunopositive (Fos-IP) neurons in spinal cord segments L3-L6 after carrageenan treatment. Carrageenan 141-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 12553707-1 2002 We have characterized segmental and laminar distribution patterns of Fos-immunopositive (Fos-IP) neurons in spinal cord segments L3-L6 after carrageenan treatment. Carrageenan 141-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 11753029-0 2002 Developmental variation in nitrous oxide-induced c-fos expression in Fischer rat spinal cord. Nitrous Oxide 27-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 12666833-0 2002 Effects of selenium and iodine on c-fos and c-jun mRNA and their protein expressions in cultured rat hippocampus cells. Selenium 11-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 12666833-0 2002 Effects of selenium and iodine on c-fos and c-jun mRNA and their protein expressions in cultured rat hippocampus cells. Iodine 24-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 12666833-1 2002 The objective of the study was to investigate the effect of selenium (Se) and iodine (I) for expressions of c-fos and c-jun mRNA and their proteins in cultured rat hippocampus cells in selenium- and iodine-containing medium. Selenium 60-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 12666833-1 2002 The objective of the study was to investigate the effect of selenium (Se) and iodine (I) for expressions of c-fos and c-jun mRNA and their proteins in cultured rat hippocampus cells in selenium- and iodine-containing medium. Iodine 78-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 12666833-1 2002 The objective of the study was to investigate the effect of selenium (Se) and iodine (I) for expressions of c-fos and c-jun mRNA and their proteins in cultured rat hippocampus cells in selenium- and iodine-containing medium. Selenium 185-193 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 12666833-1 2002 The objective of the study was to investigate the effect of selenium (Se) and iodine (I) for expressions of c-fos and c-jun mRNA and their proteins in cultured rat hippocampus cells in selenium- and iodine-containing medium. Iodine 199-205 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 12666833-3 2002 The expressions of c-fos/c-jun in cultured rat hippocampus cells (1 d, 3 d, 5 d, 7 d, and 10 d) were studied by using both in situ hybridization histochemistry and SABC immunohistochemistry techniques. sabc 164-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 11846448-0 2002 Modulation of c-myc and c-fos gene expression in regenerating rat liver by 2-mercaptopropionylglycine. Tiopronin 75-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 11930243-0 2002 [Propofol depresses c -fos expression of NOS neurons in the spinal cord of rats with inflammatory pain]. Propofol 1-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-26 11930243-1 2002 In formalin pain model, the effect of propofol on Fos expression in the spinal cord was examined by means of c -fos oncogene immunohistochemistry and NADPH-d histochemistry. Formaldehyde 3-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 11795744-0 2002 Cooling decreases fos-immunoreactivity in the rat after formalin injection. Formaldehyde 56-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 11930243-1 2002 In formalin pain model, the effect of propofol on Fos expression in the spinal cord was examined by means of c -fos oncogene immunohistochemistry and NADPH-d histochemistry. Propofol 38-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 11795744-1 2002 The effects of cooling on inflammatory response after injection of formalin in rats were examined by counting Fos-labeled cells in the dorsal horn of the spinal cord and evaluating swelling. Formaldehyde 67-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 11930243-1 2002 In formalin pain model, the effect of propofol on Fos expression in the spinal cord was examined by means of c -fos oncogene immunohistochemistry and NADPH-d histochemistry. Propofol 38-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-115 11930243-2 2002 Fos-like immunoreactive (FLI) neurons were mainly found in the ipsilateral dorsal horn after injection of formalin, some of which were FLI/NOS double-labeled neurons. Formaldehyde 106-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 11860513-5 2002 When combined, SKF 82958 and quinelorane induced c-fos expression in subthalamic neurons that was similar to that found following administration of SKF 82958 alone. 1,2,3,4-tetrahydroisoquinoline-7-sulfonamide 15-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 11849299-0 2002 The expression of Fos following kainic acid-induced seizures is age-dependent. Kainic Acid 32-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 11860513-5 2002 When combined, SKF 82958 and quinelorane induced c-fos expression in subthalamic neurons that was similar to that found following administration of SKF 82958 alone. quinelorane 29-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 11860513-9 2002 The present data show that dopamine, via D1/D5 receptors, upregulates c-fos expression in subthalamic neurons, and that the high expression of D5 receptors in this area might be involved. Dopamine 27-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 11807577-0 2002 Induction of Fos-like immunoreactivity in the basal ganglia by ether anaesthesia: effects of injections of muscimol in rostral versus caudal substantia nigra pars reticulata. Ether 63-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 11807577-1 2002 Exposing rats to ether anaesthesia for 20 min induced a massive expression of c- fos like (FL) immunoreactivity in specific regions of the brain, including the basal ganglia. Ether 17-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-84 11807577-5 2002 In experiment I, the effects of ether anaesthesia on c- fos expression in the brain were investigated. Ether 32-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-59 11807577-12 2002 Injections in both sites caused remarkable suppression of the ether-induced c- fos throughout the ipsilateral SNpr; however, the rostral injection also inhibited the expression of c- fos in the STN, EPN and GP. Ether 62-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-82 11807577-12 2002 Injections in both sites caused remarkable suppression of the ether-induced c- fos throughout the ipsilateral SNpr; however, the rostral injection also inhibited the expression of c- fos in the STN, EPN and GP. Ether 62-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 180-186 11823069-7 2002 Formalin-induced oral responses following injections into the hindpaw and the expression of Fos in the superior colliculus were virtually absent until 10 days postnatal, despite the presence of Fos-like immunoreactivity in many other structures (e.g. spinal cord, parabrachial area, periaqueductal grey). Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 11786972-6 2002 Finally, Fos expression in the nucleus tractus solitarius (NTS) was blocked by local microinjection of c-fos antisense oligonucleotides twice daily for 5 days following PVS. Oligonucleotides 119-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 11786972-6 2002 Finally, Fos expression in the nucleus tractus solitarius (NTS) was blocked by local microinjection of c-fos antisense oligonucleotides twice daily for 5 days following PVS. Oligonucleotides 119-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 11786972-9 2002 Administration of c-fos antisense oligonucleotides eliminated the hyperdynamic circulation in PVS rats, but had no effect on sham-operated controls. Oligonucleotides 34-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 11902120-5 2002 In this study we produce rat pheochromocytoma PC18 cells that overexpress c-Fos under control of the tet-inducible system. tetramethylenedisulfotetramine 101-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 11955716-7 2002 Furthermore, systemic administration of a D2 receptor antagonist, eticlopride, induced Fos immunoreactivity predominantly in globus pallidus neurons expressing preproenkephalin mRNA, while combined administration of D1 and D2 receptor agonists induced Fos predominantly in pallidal neurons lacking preproenkephalin mRNA.These results support the conclusion that preproenkephalin mRNA identifies one of the two major subpopulations of pallidal neurons. eticlopride 66-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 11955716-7 2002 Furthermore, systemic administration of a D2 receptor antagonist, eticlopride, induced Fos immunoreactivity predominantly in globus pallidus neurons expressing preproenkephalin mRNA, while combined administration of D1 and D2 receptor agonists induced Fos predominantly in pallidal neurons lacking preproenkephalin mRNA.These results support the conclusion that preproenkephalin mRNA identifies one of the two major subpopulations of pallidal neurons. eticlopride 66-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 252-255 12044461-5 2002 This was accompanied by a marked reduction in formalin-induced c-fos expression in the spinal cord. Formaldehyde 46-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 12044476-7 2002 From these findings we suggest that a down-regulation of basal Fos expression in nucleus tractus solitarii may contribute to the restoration of baroreceptor reflex sensitivity in spontaneously hypertensive rats that received antihypertensive treatment such as captopril. Captopril 260-269 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 12088752-7 2002 In contrast, under the same alpha-chloralose/urethane anesthesia, systemic infusion of sodium nitroprusside appeared to produce a hypotension and a marked increase in the density of such double c-fos and tyrosine hydroxylase expressing cells in the rostral ventrolateral medulla and the caudal medullary region surrounding the caudal part of the facial nucleus. Nitroprusside 87-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 194-199 12220572-0 2002 Lysergic acid diethylamide-induced Fos expression in rat brain: role of serotonin-2A receptors. Lysergic Acid Diethylamide 0-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 12232888-7 2002 c-Fos expression in the central nervous system was detected by immunohistochemistry by using the avidin-biotin technique. avidin-biotin 97-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 12379259-0 2002 Withdrawal duration differentially affects c-fos expression in the medial prefrontal cortex and discrete subregions of the nucleus accumbens in cocaine-sensitized rats. Cocaine 144-151 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 12379259-6 2002 The present study was designed to assess the activation of key neuronal populations in subdivisions of the accumbens and subdivisions of the medial prefrontal cortex in cocaine-sensitized rats, using the expression of the immediate early gene, c-fos, as a marker of neuronal activation. Cocaine 169-176 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 244-249 12379259-7 2002 Repeated cocaine administration resulted in robust sensitization that correlated with a significant decrease in the density of c-fos nuclei in all three subdivisions of the medial prefrontal cortex, and two subdivisions of the nucleus accumbens only in animals challenged after a 2-day withdrawal period. Cocaine 9-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 11790471-10 2002 In the lumbosacral cord, the increase in Fos expression was localized primarily to the superficial dorsal horn (SDH). sdh 112-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 11790471-11 2002 In the thoracolumbar spinal segments, Fos was induced primarily in the SDH and the area around the central canal. sdh 71-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 11823069-7 2002 Formalin-induced oral responses following injections into the hindpaw and the expression of Fos in the superior colliculus were virtually absent until 10 days postnatal, despite the presence of Fos-like immunoreactivity in many other structures (e.g. spinal cord, parabrachial area, periaqueductal grey). Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 194-197 11730984-3 2001 Neuronal excitation was visualized by in situ hybridization autoradiography (ISH) of c-fos messenger ribonucleic acid (mRNA) 45 min after intragastric (IG) administration of HCl (0.5 M; 10 ml/kg). Hydrochloric Acid 174-177 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 12020047-0 2002 Inhibition of arachidonic acid cascade attenuates the induction of c-Fos proteins by DOI, 5-HT2A/2C receptor agonist, in the rat cortex. Arachidonic Acid 14-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 12020047-4 2002 In the present study, we investigated whether arachidonic acid, a retrograde messenger, is involved in the above mechanism of c-Fos induction. Arachidonic Acid 46-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 12020047-6 2002 The inhibition of arachidonic acid cascade both at the level of phospholipase A2 (by dexamethasone, 1.5 mg/kg) or at the level of cyclooxygenases that catalyze arachidonic acid biotransformation (by indomethacin, 3 mg/kg), decreased the number of c-Fos immunopositive cells after induction by DOI (8 mg/kg). Arachidonic Acid 18-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 247-252 12020047-7 2002 Our results suggest that arachidonic acid cascade may be involved in the induction of c-Fos proteins by DOI in the rat parietal cortex. Arachidonic Acid 25-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 11788132-0 2002 CO(2)-induced c-Fos expression in hypothalamic vasopressin containing neurons. co(2) 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 11788132-4 2002 A marked increase in c-Fos positive cells was induced after 2 h of breathing a gas mixture with elevated CO(2) (5% CO(2), 21% O(2) and 74% N(2), or 1 h following breathing of 12% CO(2,) 21% O(2,) and 67% N(2)). co(2) 105-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 11788132-4 2002 A marked increase in c-Fos positive cells was induced after 2 h of breathing a gas mixture with elevated CO(2) (5% CO(2), 21% O(2) and 74% N(2), or 1 h following breathing of 12% CO(2,) 21% O(2,) and 67% N(2)). co(2) 115-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 11788132-4 2002 A marked increase in c-Fos positive cells was induced after 2 h of breathing a gas mixture with elevated CO(2) (5% CO(2), 21% O(2) and 74% N(2), or 1 h following breathing of 12% CO(2,) 21% O(2,) and 67% N(2)). Nitrogen 139-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 11788132-4 2002 A marked increase in c-Fos positive cells was induced after 2 h of breathing a gas mixture with elevated CO(2) (5% CO(2), 21% O(2) and 74% N(2), or 1 h following breathing of 12% CO(2,) 21% O(2,) and 67% N(2)). co(2 105-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 11788132-4 2002 A marked increase in c-Fos positive cells was induced after 2 h of breathing a gas mixture with elevated CO(2) (5% CO(2), 21% O(2) and 74% N(2), or 1 h following breathing of 12% CO(2,) 21% O(2,) and 67% N(2)). Nitrogen 204-205 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 11739600-5 2001 In intact rats, CREB antisense reduced both basal and cocaine-induced expression of c-Fos, FosB/DeltaFosB, and prodynorphin mRNA. Cocaine 54-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 11730984-7 2001 Subcutaneous administration of the NK3 receptor antagonist SB-222,200 (20 mg/kg) reduced the c-fos mRNA response in AP and SFO and enhanced it in Hb. sb-222 59-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 11746441-1 2001 Kainic acid injected in vivo into adult rats evokes the expression of the immediate early gene c-fos in the dentate gyrus and associated structures before a seizure occurs and in these and additional regions after a single motor seizure. Kainic Acid 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 11746441-4 2001 Given the early Fos-labeling of these cells, we suggest they are associated with the hippocampal EEG events also seen at this stage of the effects of kainic acid. Kainic Acid 150-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 11720724-3 2001 VCS also induces the expression of Fos, the protein product of the immediate early gene c-fos, which has been used as a marker for neurons that are responsive to mating stimuli. VCP 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 11720724-3 2001 VCS also induces the expression of Fos, the protein product of the immediate early gene c-fos, which has been used as a marker for neurons that are responsive to mating stimuli. VCP 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 11720724-4 2001 Because VCS induces the release of dopamine in the forebrain, as well as phosphorylation of DARPP-32, a phosphoprotein associated with activation of the D(1) subtype of dopamine receptor, we tested the hypothesis that VCS induces Fos expression by acting on the D(1) class of dopamine receptors. VCP 8-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 230-233 11720724-5 2001 Injection of SCH 23390, an antagonist of the D(1) class of dopamine receptors, virtually eliminated VCS-induced Fos expression without affecting constitutive levels of Fos-Immunoreactivity (Fos-IR) in all brain areas in which VCS induced Fos expression. SCH 23390 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 11726797-1 2001 Systemic administration of a dopamine D2 receptor blocker, haloperidol, but not vehicle, significantly increased the number of c-Fos-immunoreactive neurons in the globus pallidus (GP) in rats. Haloperidol 59-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 11726797-3 2001 Lesioning of the subthalamic nucleus (STN) by local injection of ibotenic acid inhibited the haloperidol-induced c-Fos expression in the GP neurons, suggesting that the activation of GP neurons is a result of increased excitatory drives from the STN. Ibotenic Acid 65-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 11726797-3 2001 Lesioning of the subthalamic nucleus (STN) by local injection of ibotenic acid inhibited the haloperidol-induced c-Fos expression in the GP neurons, suggesting that the activation of GP neurons is a result of increased excitatory drives from the STN. Haloperidol 93-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 11709423-6 2001 This depolarization-induced increase in [Ca(2+)](i) was associated with increased activation of the transcription factor, cAMP response element binding protein, and increased expression of the immediate early gene c-fos, both of which are reversed by acute exposure to the voltage-dependent Ca(2+) channel blocker nisoldipine. Nisoldipine 314-325 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 214-219 11724746-0 2001 The effects of dentate granule cell destruction on behavioural activity and Fos protein expression induced by systemic methamphetamine in rats. Methamphetamine 119-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 11724746-8 2001 Bilaterally lesioned animals, when administered saline and having undergone an immunohistological examination, showed a marked increase in Fos expression in both sides of the nucleus accumbens. Sodium Chloride 48-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 11724746-9 2001 Bilaterally lesioned animals administered methamphetamine showed a marked increase in Fos expression in the right and left sides of all regions tested. Methamphetamine 42-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 11724746-10 2001 Unilaterally lesioned animals administered methamphetamine showed a significant and bilateral enhancement in Fos expression in the medial prefrontal and cingulate cortices, and a marked and unilateral (ipsilateral to the lesioned side) enhancement of Fos protein in the piriform cortex, dorsal striatum, and nucleus accumbens. Methamphetamine 43-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-112 11724746-10 2001 Unilaterally lesioned animals administered methamphetamine showed a significant and bilateral enhancement in Fos expression in the medial prefrontal and cingulate cortices, and a marked and unilateral (ipsilateral to the lesioned side) enhancement of Fos protein in the piriform cortex, dorsal striatum, and nucleus accumbens. Methamphetamine 43-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 251-254 11843868-6 2001 In addition, the ventrolateral periaqueductal grey, the intralaminar thalamic and various hypothalamic areas, showed an enhanced Fos expression after the intracisternal administration of capsaicin. Capsaicin 187-196 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-132 11720724-5 2001 Injection of SCH 23390, an antagonist of the D(1) class of dopamine receptors, virtually eliminated VCS-induced Fos expression without affecting constitutive levels of Fos-Immunoreactivity (Fos-IR) in all brain areas in which VCS induced Fos expression. VCP 100-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 11720724-7 2001 Therefore, the results are consistent with the idea that VCS induces dopamine release, causing activation of D(1) dopamine receptors, which in turn, results in neuronal response, as seen by both Fos and egr-1 expression. VCP 57-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-198 11720724-7 2001 Therefore, the results are consistent with the idea that VCS induces dopamine release, causing activation of D(1) dopamine receptors, which in turn, results in neuronal response, as seen by both Fos and egr-1 expression. Dopamine 69-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-198 11720732-0 2001 The distribution of cannabinoid-induced Fos expression in rat brain: differences between the Lewis and Wistar strain. Cannabinoids 20-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 11737151-8 2001 We discovered that another portion of DIF-1 cytotoxicity is independent of the NMDA receptor; that is, coaddition of DIF-1 and MK801 induced dendritic beading and increased expression of the immediate early genes c-fos and zif/268. Dizocilpine Maleate 127-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-218 11760011-1 2001 Maternal treatment with cocaine or a D1-dopamine receptor agonist induces c-fos gene expression in the fetal suprachiasmatic nuclei (SCN). Cocaine 24-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 11760011-2 2001 Other treatments that induce c-fos expression in the fetal SCN include caffeine and nicotine. Caffeine 71-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 11760011-2 2001 Other treatments that induce c-fos expression in the fetal SCN include caffeine and nicotine. Nicotine 84-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 11752896-0 2001 Changes in Fos expression in various brain regions during deoxycorticosterone acetate treatment: relation to salt appetite, vasopressin mRNA and the mineralocorticoid receptor. Desoxycorticosterone Acetate 58-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 11752896-0 2001 Changes in Fos expression in various brain regions during deoxycorticosterone acetate treatment: relation to salt appetite, vasopressin mRNA and the mineralocorticoid receptor. Salts 109-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 11752896-5 2001 In most of these areas, increased Fos expression was also observed early (2 h) after a single DOCA injection, well before salt appetite develops. Desoxycorticosterone Acetate 94-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 11752896-9 2001 The present demonstration of Fos activation, in conjunction with differential expression of MR and stimulation of AVP mRNA, suggests that a neuroanatomical pathway comprising the AMYG, osmosensitive brain regions and magnocellular nuclei becomes activated during DOCA effects on salt appetite. Desoxycorticosterone Acetate 263-267 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 11752896-9 2001 The present demonstration of Fos activation, in conjunction with differential expression of MR and stimulation of AVP mRNA, suggests that a neuroanatomical pathway comprising the AMYG, osmosensitive brain regions and magnocellular nuclei becomes activated during DOCA effects on salt appetite. Salts 279-283 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 11716816-0 2001 Environmental context modulates the ability of cocaine and amphetamine to induce c-fos mRNA expression in the neocortex, caudate nucleus, and nucleus accumbens. Cocaine 47-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 11716816-0 2001 Environmental context modulates the ability of cocaine and amphetamine to induce c-fos mRNA expression in the neocortex, caudate nucleus, and nucleus accumbens. Amphetamine 59-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 11716816-2 2001 The purpose of the present experiment was 2-fold: to determine (1) whether environmental novelty has a similar effect on the ability of cocaine to induce c-fos mRNA, and (2) whether this effect is seen in neurologically-intact rats (in previous experiments we studied the intact hemisphere of rats with a unilateral 6-OHDA lesion). Cocaine 136-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 11716816-3 2001 In the dorsal portion of the caudate putamen, core and shell of the nucleus accumbens, and in several cortical regions, both amphetamine (1.5 mg/kg) and cocaine (15 mg/kg) induced higher levels of c-fos mRNA expression when administered in a novel environment, relative to when they were administered in the home cage. Amphetamine 125-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 197-202 11716816-3 2001 In the dorsal portion of the caudate putamen, core and shell of the nucleus accumbens, and in several cortical regions, both amphetamine (1.5 mg/kg) and cocaine (15 mg/kg) induced higher levels of c-fos mRNA expression when administered in a novel environment, relative to when they were administered in the home cage. Cocaine 153-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 197-202 11716842-3 2001 In the present study, we performed c-Fos immunohistochemistry in the dopaminergic terminal regions of adolescent rat brain after the intraperitoneal administration of dextromethorphan at different doses (0, 10, 20, and 40 mg/kg), and also examined the effects on nocturnal behavior. Dextromethorphan 167-183 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 11716842-4 2001 The results showed that dextromethorphan increased c-Fos expression dose dependently in the anterior cingulate cortex, caudate putamen, nucleus accumbens, and central amygdala. Dextromethorphan 24-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 11707641-0 2001 Regional specificity of ethanol and NMDA action in brain revealed with FOS-like immunohistochemistry and differential routes of drug administration. Ethanol 24-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 11707641-0 2001 Regional specificity of ethanol and NMDA action in brain revealed with FOS-like immunohistochemistry and differential routes of drug administration. N-Methylaspartate 36-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 11707641-3 2001 METHODS: The induction of Fos-like immunoreactivity (Fos-LI) in 38 regions of the rat brain was measured 2 hr after treatment with NMDA, EtOH, or both. N-Methylaspartate 131-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 11707641-3 2001 METHODS: The induction of Fos-like immunoreactivity (Fos-LI) in 38 regions of the rat brain was measured 2 hr after treatment with NMDA, EtOH, or both. N-Methylaspartate 131-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 11707641-3 2001 METHODS: The induction of Fos-like immunoreactivity (Fos-LI) in 38 regions of the rat brain was measured 2 hr after treatment with NMDA, EtOH, or both. Ethanol 137-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 11707641-3 2001 METHODS: The induction of Fos-like immunoreactivity (Fos-LI) in 38 regions of the rat brain was measured 2 hr after treatment with NMDA, EtOH, or both. Ethanol 137-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 11707641-6 2001 RESULTS: For the 38 forebrain regions examined, ip and iv NMDA significantly induced Fos-LI in 13 and 32 regions, respectively. N-Methylaspartate 58-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 11707641-8 2001 For EtOH, prominent Fos-LI induction was found in the central amygdala, dorsolateral bed nucleus of the stria terminalis, Edinger-Westphal nucleus, and paraventricular hypothalamus. Ethanol 4-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 11707641-9 2001 Despite ip and ig EtOH induction of Fos-LI in these regions, the major effect of EtOH was to block NMDA-induced Fos-LI in 8 of 13 (ip) and 27 of 32 (ig) of the NMDA-positive regions, respectively, including retrosplenial, cingulate, and medial prefrontal cortices, central amygdala, and taenia tecta. Ethanol 81-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 11707641-9 2001 Despite ip and ig EtOH induction of Fos-LI in these regions, the major effect of EtOH was to block NMDA-induced Fos-LI in 8 of 13 (ip) and 27 of 32 (ig) of the NMDA-positive regions, respectively, including retrosplenial, cingulate, and medial prefrontal cortices, central amygdala, and taenia tecta. N-Methylaspartate 99-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 11668041-3 2001 The neurochemical identity of the neurons activated by the HCl challenge was determined by colocalizing c-Fos with a marker for excitatory pathways, choline acetyltransferase (ChAT), and a marker for inhibitory pathways, nitric oxide synthase (NOS). Hydrochloric Acid 59-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 11668041-5 2001 Treatment with 0.35, 0.5, and 0.7 M HCl induced c-Fos in 8%, 56%, and 64%, respectively, of NOS-positive but not ChAT-positive neurons. Hydrochloric Acid 36-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 11668041-6 2001 c-Fos was also seen in glial cells of HCl-treated rats, whereas in saline-treated animals c-Fos was absent from the myenteric plexus. Hydrochloric Acid 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 11698319-0 2001 Intrathecal pre-administration of fentanyl effectively suppresses formalin evoked c-Fos expression in spinal cord of rat. Fentanyl 34-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 11698319-0 2001 Intrathecal pre-administration of fentanyl effectively suppresses formalin evoked c-Fos expression in spinal cord of rat. Formaldehyde 66-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 11698319-9 2001 Pretreatment with fentanyl showed the most profound suppression of c-Fos expression (P <0.01). Fentanyl 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 11698319-11 2001 Naloxone reversed the action of fentanyl on c-Fos activity. Naloxone 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 11698319-11 2001 Naloxone reversed the action of fentanyl on c-Fos activity. Fentanyl 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 12086906-6 2001 CCh induced time-and dose-dependent increases in the c-fos and c-jun early-response genes, which were blocked by m1 and m3 inhibition but not by m2 inhibition. Carbachol 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 11606657-5 2001 The number of Fos-positive cells after acetic acid administration were significantly reduced in the L6 spinal cord from antisense-treated animals, compared with mismatch ODN-treated animals. Acetic Acid 39-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 11698518-6 2001 Compared with saline infusion, acetic acid infusion caused a significant increase in c-fos expression at L(1) and L(6) of the spinal cord, and HGN transection significantly reduced c-fos expression in the dorsal horn of the spinal cord at L(1) but not at L(6). Acetic Acid 31-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 11754968-3 2001 Few cells with Fos-LI were found in the CNS 90 min after icv administration of saline. Sodium Chloride 79-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-18 11754968-4 2001 Fos-LI was also detected in the various hypothalamic areas after icv administration of PAMP. pamp 87-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 11702093-3 2001 However, anatomical studies of Fos expression suggest that nicotine targets primarily non-cholinergic neurons in the PPTg, especially GABAergic and glutamatergic neurons. Nicotine 59-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 21207693-7 2001 RESULTS: NADPH-diaphorase positive neurons were densely distributed in CGLD, CGLV and DR. NADPH-d, Fos and NADPH/Fos double-labeled neurons appeared in CGLD, CGLV and DR after injection of formalin in the rat hindpaw. NADP 9-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-116 11698063-0 2001 Characterization of the signal transduction pathways mediating morphine withdrawal-stimulated c-fos expression in hypothalamic nuclei. Morphine 63-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 11698063-2 2001 We have shown previously that acute administration of morphine induces the expression of Fos in hypothalamic nuclei associated with control of the hypothalamus-pituitary-adrenocortex axis, such as the paraventricular nucleus and the supraoptic nucleus. Morphine 54-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 11698063-3 2001 In the current study, we examined the role of protein kinase A, protein kinase C and Ca2+ entry through L-type Ca2+ channels in naloxone-precipitated Fos expression in the paraventricular and supraoptic nuclei. Naloxone 128-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-153 11698063-5 2001 However, when opioid withdrawal was precipitated with naloxone a dramatic increase in Fos immunoreactivity was observed in the parvocellular division of the paraventricular nucleus and in the supraoptic nucleus. Naloxone 54-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 11698063-9 2001 Furthermore, chronic infusion of the selective L-type Ca2+ channel antagonist, nimodipine, significantly inhibited the enhancement of Fos induction in the paraventricular and supraoptic nuclei from morphine-withdrawn animals. Nimodipine 79-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-137 11698063-9 2001 Furthermore, chronic infusion of the selective L-type Ca2+ channel antagonist, nimodipine, significantly inhibited the enhancement of Fos induction in the paraventricular and supraoptic nuclei from morphine-withdrawn animals. Morphine 198-206 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-137 11698063-10 2001 Taken together, these data might indicate that protein kinase A activity is necessary for the expression of Fos during morphine withdrawal and that an up-regulated Ca2+ system might contribute to the activation of Fos. Morphine 119-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 11597596-4 2001 Rats in which ibotenic acid had been injected into the BNST showed attenuated expression of c-Fos in the hypothalamic paraventricular nucleus (PVN) and blunted elevation of plasma adrenocorticotropic hormone (ACTH) after microinjection of neostigmine into the hippocampus compared with rats in which saline had been injected into the BNST. Ibotenic Acid 14-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 11595203-3 2001 Gastrointestinal sensors were stimulated by gastric balloon distension or by intraduodenal infusion of either linoleic acid or glucose in chronically catheterized, non-anesthetized rats, leading to activation of second order neurons in the NTS as detected by c-Fos immunohistochemistry. Glucose 127-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 259-264 11641538-1 2001 Following kainate (KA)-induced epilepsy, rat hippocampal neurons strongly express immediate early gene (IEG) products, i.e., c-FOS and c-JUN, and neural stress protein, HSP72. Kainic Acid 10-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 11689187-2 2001 Previously, by studying c-fos expression, we showed that the striatum was a possible target region for the antimanic effects of lithium salt. lithium salt 128-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 11551840-2 2001 Subcutaneous nicotine injection moderately increased Fos expression in the principal ganglionic cells of the CG (17 +/- 4 Fos+ per mm(2), approximately 12% of all principal CG cells), whereas subcutaneous saline had no effect (0 +/- 0 Fos+ per mm(2); n = 7; P < 0.01). Nicotine 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 11551840-2 2001 Subcutaneous nicotine injection moderately increased Fos expression in the principal ganglionic cells of the CG (17 +/- 4 Fos+ per mm(2), approximately 12% of all principal CG cells), whereas subcutaneous saline had no effect (0 +/- 0 Fos+ per mm(2); n = 7; P < 0.01). Nicotine 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 11551840-2 2001 Subcutaneous nicotine injection moderately increased Fos expression in the principal ganglionic cells of the CG (17 +/- 4 Fos+ per mm(2), approximately 12% of all principal CG cells), whereas subcutaneous saline had no effect (0 +/- 0 Fos+ per mm(2); n = 7; P < 0.01). Nicotine 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 11551840-3 2001 Greater Fos expression was obtained by applying nicotine topically to the CG (71 +/- 8 Fos+ per mm(2); 52% of all principal CG cells, n = 5; P < 0.01 vs. topical saline, n = 4) and by preganglionic nerve stimulation (126 +/- 9 Fos+ per mm(2); 94% of all principal CG cells, n = 11; P < 0.01 vs. nerve isolation, n = 7). Nicotine 48-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-11 11551840-3 2001 Greater Fos expression was obtained by applying nicotine topically to the CG (71 +/- 8 Fos+ per mm(2); 52% of all principal CG cells, n = 5; P < 0.01 vs. topical saline, n = 4) and by preganglionic nerve stimulation (126 +/- 9 Fos+ per mm(2); 94% of all principal CG cells, n = 11; P < 0.01 vs. nerve isolation, n = 7). Nicotine 48-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 11551840-3 2001 Greater Fos expression was obtained by applying nicotine topically to the CG (71 +/- 8 Fos+ per mm(2); 52% of all principal CG cells, n = 5; P < 0.01 vs. topical saline, n = 4) and by preganglionic nerve stimulation (126 +/- 9 Fos+ per mm(2); 94% of all principal CG cells, n = 11; P < 0.01 vs. nerve isolation, n = 7). Nicotine 48-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 11551840-3 2001 Greater Fos expression was obtained by applying nicotine topically to the CG (71 +/- 8 Fos+ per mm(2); 52% of all principal CG cells, n = 5; P < 0.01 vs. topical saline, n = 4) and by preganglionic nerve stimulation (126 +/- 9 Fos+ per mm(2); 94% of all principal CG cells, n = 11; P < 0.01 vs. nerve isolation, n = 7). Sodium Chloride 165-171 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-11 11551840-4 2001 Moderate Fos expression was also observed in the CG after intraperitoneal 2-deoxy-D-glucose (2DG) injection (21 +/- 2 Fos+ per mm(2); 16% of all principal CG cells, n = 5; P < 0.01 vs. saline ip) or insulin injection (16 +/- 2 Fos+ per mm(2); 12% of all principal CG cells, n = 6; P < 0.01 vs. saline ip). Deoxyglucose 74-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 11551840-4 2001 Moderate Fos expression was also observed in the CG after intraperitoneal 2-deoxy-D-glucose (2DG) injection (21 +/- 2 Fos+ per mm(2); 16% of all principal CG cells, n = 5; P < 0.01 vs. saline ip) or insulin injection (16 +/- 2 Fos+ per mm(2); 12% of all principal CG cells, n = 6; P < 0.01 vs. saline ip). Deoxyglucose 93-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 11551840-4 2001 Moderate Fos expression was also observed in the CG after intraperitoneal 2-deoxy-D-glucose (2DG) injection (21 +/- 2 Fos+ per mm(2); 16% of all principal CG cells, n = 5; P < 0.01 vs. saline ip) or insulin injection (16 +/- 2 Fos+ per mm(2); 12% of all principal CG cells, n = 6; P < 0.01 vs. saline ip). Deoxyglucose 93-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 11551840-4 2001 Moderate Fos expression was also observed in the CG after intraperitoneal 2-deoxy-D-glucose (2DG) injection (21 +/- 2 Fos+ per mm(2); 16% of all principal CG cells, n = 5; P < 0.01 vs. saline ip) or insulin injection (16 +/- 2 Fos+ per mm(2); 12% of all principal CG cells, n = 6; P < 0.01 vs. saline ip). Deoxyglucose 93-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 11551840-5 2001 Furthermore, Fos expression induced by 2DG was dose and time dependent. Deoxyglucose 39-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 11574375-0 2001 A prostaglandin E2 receptor subtype EP1 receptor antagonist (ONO-8711) reduces hyperalgesia, allodynia, and c-fos gene expression in rats with chronic nerve constriction. ONO 8711 61-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 11600104-0 2001 Olanzapine activates the rat locus coeruleus: in vivo electrophysiology and c-Fos immunoreactivity. Olanzapine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 11600104-4 2001 The effects of olanzapine and clozapine on c-Fos expression in the LC, nucleus subcoeruleus part alpha (SubCA), and nucleus A5 (A5) were studied by immunohistochemistry. Clozapine 30-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 11600104-6 2001 Induction of c-Fos expression in the LC by olanzapine and clozapine was confirmed and was also found in the SubCA, but not in A5. Olanzapine 43-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 11600104-6 2001 Induction of c-Fos expression in the LC by olanzapine and clozapine was confirmed and was also found in the SubCA, but not in A5. Clozapine 58-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 11703467-5 2001 Furthermore, whisker stimulation-evoked c-fos and zif 268 expression in the barrel cortex ipsilateral to the lesion was also attenuated by acute dopamine depletion. Dopamine 145-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 11494404-4 2001 Additionally, in prenatal cocaine-exposed rats dopaminergic neurons in the ventral, midline A10, and lateral A9 regions demonstrated a hyperreactivity to environmental stress, as measured by activation of the immediate-early gene, Fos. Cocaine 26-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 231-234 11568155-6 2001 Histochemical and immunocytochemical data showed that the right FNr contained clustered HRP-labeled neurons, most of which were double labeled with c-Fos immunoreactivity in both electrically and CO(2)-stimulated rats. co(2) 196-201 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 11587712-2 2001 Withdrawal precipitated with a peripherally acting quaternary opioid antagonist (naloxone methiodide) increased Fos expression but caused a more restricted pattern of neuronal activation than systemic withdrawal (precipitated with naloxone which enters the brain). N-methylnaloxone 81-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 11587712-2 2001 Withdrawal precipitated with a peripherally acting quaternary opioid antagonist (naloxone methiodide) increased Fos expression but caused a more restricted pattern of neuronal activation than systemic withdrawal (precipitated with naloxone which enters the brain). Naloxone 81-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 11587713-0 2001 Modulation of primary afferent-mediated neurotransmission and Fos expression by glutamate uptake inhibition in rat spinal neurones in vitro. Glutamic Acid 80-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 11587713-7 2001 L-PDC (1mM) or L-AP4 (30 microM) reduced afferent-evoked dorsal horn Fos expression, this effect was reversed by CPPG. l-pdc 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 11587713-7 2001 L-PDC (1mM) or L-AP4 (30 microM) reduced afferent-evoked dorsal horn Fos expression, this effect was reversed by CPPG. 2-amino-4-phosphono-propinate 15-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 11833421-6 2001 (4) Fos protein expression induced by renal ischemia was significantly inhibited by pretreatment with 8-PT (P < 0.05). 8-phenyltheophylline 102-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-7 11578603-3 2001 Following induction of CCI, a very large number of Fos-IR neurons appeared in the spinal dorsal horn, but a significant number of Fos-IR neurons were also observed in the contralateral dorsal horn where primary afferents of the injured sciatic nerve rarely project. CCI 23-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 11578603-6 2001 The ratio of the number of Fos-IR neurons in the ipsilateral dorsal horn to the contralateral dorsal horn, however, had its peak level 3 days following CCI (3.1-fold increase compared to the contralateral dorsal horn). CCI 152-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 11578603-9 2001 The present results indicate that the induction of Fos-IR neurons in the dorsal horn caused by CCI is biphasic and reaches its maximal level on PO 3d, near the time of hyperalgesia onset. CCI 95-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 11589989-0 2001 Dynamics of c-fos and ICER mRNA expression in rat forebrain following lithium chloride injection. Lithium Chloride 70-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 11589989-2 2001 LiCl administration in rats has been correlated with activation of c-fos and cAMP-mediated gene transcription in many brain regions; however, little is known about the timing or duration of gene activation. Lithium Chloride 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 11589989-3 2001 We hypothesized that c-fos gene transcription is rapidly stimulated by LiCl, followed later by the expression of the inducible cAMP early repressor (ICER) transcription factor, a negative modulator of cAMP-mediated gene transcription. Cyclic AMP 127-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 11589989-3 2001 We hypothesized that c-fos gene transcription is rapidly stimulated by LiCl, followed later by the expression of the inducible cAMP early repressor (ICER) transcription factor, a negative modulator of cAMP-mediated gene transcription. Cyclic AMP 201-205 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 11588591-2 2001 injection of nitric oxide (NO)-related pharmacological agents affect Fos activation in the hippocampus and cortex of lactating rats. Nitric Oxide 13-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 11588591-5 2001 Urethane anaesthesia abolished Fos expression in the cortex, but not in the hippocampus following simultaneous SNP injection and resumption of suckling. Urethane 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 11494405-0 2001 Maturational increases in c-fos expression in the ascending dopamine systems. Dopamine 60-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 11494405-5 2001 When expressed as a percentage of vehicle for each age, amphetamine-induced effects on c-fos immunoreactivity were higher at 21 days of age compared with the effects at 35 and 60 days of age in the nucleus accumbens core and shell, striatum, and prefrontal cortex. Amphetamine 56-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 11543769-0 2001 Induction of Fos-immunostaining by nicotine and nicotinic receptor antagonists in rat brain. Nicotine 35-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 11557310-9 2001 The results indicated significant induction of JNK-1 and c-fos proteins in the ischemic/reperfused myocardium, which was inhibited by the proanthocyanidin extract. proanthocyanidin 138-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 11585578-7 2001 The activation of c-fos by magnetic stimulation was inhibited by the sodium-channel blocker tetrodotoxin (TTX) (10 microM). Tetrodotoxin 92-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 11585578-7 2001 The activation of c-fos by magnetic stimulation was inhibited by the sodium-channel blocker tetrodotoxin (TTX) (10 microM). Tetrodotoxin 106-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 11532430-8 2001 DEX treatment significantly enhanced the number of Fos positive nuclei in Laminae I-III by 4 h after transection, although the response was not maintained by intensive steroid treatment when tested at 48 h after SCI. Dexamethasone 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 11553360-1 2001 In previous studies using Fos expression as a marker of neuronal activation, we showed that nitrous oxide (N(2)O) activates bulbospinal noradrenergic neurons in rats and that destruction of these neuronal pathways leads to loss of N(2)O antinociceptive action. Nitrous Oxide 92-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 11553360-1 2001 In previous studies using Fos expression as a marker of neuronal activation, we showed that nitrous oxide (N(2)O) activates bulbospinal noradrenergic neurons in rats and that destruction of these neuronal pathways leads to loss of N(2)O antinociceptive action. Nitrous Oxide 107-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 11506987-0 2001 Estradiol treatment increases feeding-induced c-Fos expression in the brains of ovariectomized rats. Estradiol 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 11506987-6 2001 Feeding increased the number of c-Fos-positive cells in the NTS, the paraventricular nucleus of the hypothalamus (PVN), and the central nucleus of the amygdala (CeA) in oil-treated rats. Oils 169-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 11506987-8 2001 Estradiol treatment also increased feeding-induced c-Fos in the PVN and CeA. Estradiol 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 11564650-14 2001 Immunohistochemical studies show Fos expression in the dorsal vagal complex (DVC) of the brainstem of 2-DG-treated animals. Deoxyglucose 102-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 11564650-16 2001 increased the number of Fos-immunoreactive cells induced by 2-DG, both in the nucleus tractus solitarii (NTS) and in the dorsal motor nucleus (DMN) of the DVC. Deoxyglucose 60-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 11564650-17 2001 Pre-treatment with L-NAME prevented the increase in Fos expression induced by endotoxin in both nuclei. NG-Nitroarginine Methyl Ester 19-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 11521152-2 2001 Pretreatment with the c-fos antisense ODN significantly decreased the CFA-induced expression of c-Fos protein dose-dependently in ipsilateral laminae I/II (LI/II) of the dorsal horn (mean +/- SEM per section: 10 nM ODN, 43.9+/-1.3; 25 nM ODN, 19.4+/-4.1) compared with pretreatment with the mismatch ODN (63.6+/-2.9; 60.6+/-4.0) or saline (56.6+/-5.5). Sodium Chloride 332-338 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 11521152-2 2001 Pretreatment with the c-fos antisense ODN significantly decreased the CFA-induced expression of c-Fos protein dose-dependently in ipsilateral laminae I/II (LI/II) of the dorsal horn (mean +/- SEM per section: 10 nM ODN, 43.9+/-1.3; 25 nM ODN, 19.4+/-4.1) compared with pretreatment with the mismatch ODN (63.6+/-2.9; 60.6+/-4.0) or saline (56.6+/-5.5). Sodium Chloride 332-338 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 11521152-3 2001 Animals pre-treated with 25 nM of the c-fos antisense ODN significantly increased the withdrawal latency to the noxious thermal stimulation (63.0-70.5%; compared with contralateral to the CFA injection) compared with animals pretreated with mismatch ODN (28.5-42.6%) or saline (26.4-45.3%) from 0 to 5 h after unilateral injection of CFA into the hind footpad. Sodium Chloride 270-276 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 11555177-4 2001 The present study shows that the stimulation of cultured pinealocytes by 1 microM epinephrine (an alpha- and beta-adrenergic agonist) for 2 h increased the number of c-fos immunoreactive (IR) cells, and that this stimulatory effect was abolished by adding 10 microM prazosin (an alpha-adrenergic antagonist) to the culture medium. Epinephrine 82-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-171 11543769-2 2001 Acute nicotine elevated Fos-like immunostaining (Fos IS) significantly in all studied areas except the medial prefrontal cortex. Nicotine 6-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 11543769-2 2001 Acute nicotine elevated Fos-like immunostaining (Fos IS) significantly in all studied areas except the medial prefrontal cortex. Nicotine 6-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 11543769-3 2001 Nicotine increased the Fos IS in cortical, limbic and hypothalamic areas by 2-10-fold, and in the interpeduncular nucleus as well as in the visual areas the increases were 15-150-fold. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 11543769-8 2001 The efficacy of nAChR antagonists in blocking nicotine"s effects on Fos IS varied noticeably with respect to region and antagonist, and the combined effect of nicotine+antagonist did not exceed that of either treatment alone. Nicotine 46-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 11543769-9 2001 Mecamylamine and DHE significantly reduced nicotine-induced Fos IS in most of the studied areas, and MLA only in two areas. Mecamylamine 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 11543769-9 2001 Mecamylamine and DHE significantly reduced nicotine-induced Fos IS in most of the studied areas, and MLA only in two areas. Dihydro-beta-Erythroidine 17-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 11543769-9 2001 Mecamylamine and DHE significantly reduced nicotine-induced Fos IS in most of the studied areas, and MLA only in two areas. Nicotine 43-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 11543769-10 2001 Thus, nAChRs seem to mediate the effects of nicotine on Fos IS, and the differences in the effects of the antagonists studied suggest that more than one subtype of nAChRs are involved. Nicotine 44-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 11561259-7 2001 The number of c-fos-positive cells increased as the formalin concentration increased. Formaldehyde 52-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 11561259-8 2001 Halothane inhalation affected the results of both the behavior and the c-fos immunoreactivity, especially in the 10% formalin (3.7% formaldehyde) group. Halothane 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 11561259-8 2001 Halothane inhalation affected the results of both the behavior and the c-fos immunoreactivity, especially in the 10% formalin (3.7% formaldehyde) group. Formaldehyde 117-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 11561718-0 2001 Differential effects of forskolin and phobol 12-myristate-13-acetate on the c-fos and c-jun mRNA expression in rat C6 glioma cells. Colforsin 24-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 11561718-0 2001 Differential effects of forskolin and phobol 12-myristate-13-acetate on the c-fos and c-jun mRNA expression in rat C6 glioma cells. phobol 12-myristate-13-acetate 38-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 11561718-1 2001 The effects of forskolin (FSK) and phobol 12-myristate-13-acetate (PMA) on c-fos and c-jun mRNA expressions in rat C6 glioma cells were studied. Colforsin 26-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 11561718-1 2001 The effects of forskolin (FSK) and phobol 12-myristate-13-acetate (PMA) on c-fos and c-jun mRNA expressions in rat C6 glioma cells were studied. phobol 12-myristate-13-acetate 35-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 11561718-1 2001 The effects of forskolin (FSK) and phobol 12-myristate-13-acetate (PMA) on c-fos and c-jun mRNA expressions in rat C6 glioma cells were studied. Tetradecanoylphorbol Acetate 67-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 11561718-2 2001 Both FSK and PMA increased the c-fos mRNA level. Colforsin 5-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 11561718-4 2001 The elevated c-fos mRNA level, induced by FSK or PMA, was significantly inhibited by dexamethasone (DEX). Colforsin 42-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 11561718-4 2001 The elevated c-fos mRNA level, induced by FSK or PMA, was significantly inhibited by dexamethasone (DEX). Dexamethasone 85-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 11561718-4 2001 The elevated c-fos mRNA level, induced by FSK or PMA, was significantly inhibited by dexamethasone (DEX). Dexamethasone 100-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 11561718-6 2001 Cycloheximide (CHX) caused a superinduction of the FSK- or PMA-induced c-fos mRNA level. Cycloheximide 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 11561718-6 2001 Cycloheximide (CHX) caused a superinduction of the FSK- or PMA-induced c-fos mRNA level. Cycloheximide 15-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 11561718-6 2001 Cycloheximide (CHX) caused a superinduction of the FSK- or PMA-induced c-fos mRNA level. Colforsin 51-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 11561718-6 2001 Cycloheximide (CHX) caused a superinduction of the FSK- or PMA-induced c-fos mRNA level. Tetradecanoylphorbol Acetate 59-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 11561718-12 2001 In addition, DEX appears to have a selective inhibitory action against c-fos, but not c-jun, -mRNA expression that is regulated by PKA and PKC. Dexamethasone 13-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 11561718-13 2001 On-going protein synthesis inhibition is required for the superinduction of the c-fos expression that is induced by PMA, or FSK and the PMA-induced c-jun mRNA level. Colforsin 124-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 11769611-11 2001 Treatment with BDM induced an earlier expression of c-myc mRNA at 6 hours, and increased the gene and protein expressions of c-fos and bFGF. diacetylmonoxime 15-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 11769627-1 2001 OBJECTIVES: This study was conducted to explore effects of selenium on hepatocellular protooncogene c-myc, c-fos and c-jun expression induced by cadmium in rats. Selenium 59-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 11769627-1 2001 OBJECTIVES: This study was conducted to explore effects of selenium on hepatocellular protooncogene c-myc, c-fos and c-jun expression induced by cadmium in rats. Cadmium 145-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 11769627-5 2001 RESULTS: The results showed that cadmium chloride at doses of 5, 10 or 20 mumol/kg, significantly induced proto-oncogene c-myc, c-fos and c-jun expression, and when sodium selenite at the dose of 5 mumol/kg was given at the time, the effect of cadmium chloride on hepatocellular protooncogene c-myc, c-fos and c-jun expression was inhibited. Cadmium Chloride 33-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-133 11769627-5 2001 RESULTS: The results showed that cadmium chloride at doses of 5, 10 or 20 mumol/kg, significantly induced proto-oncogene c-myc, c-fos and c-jun expression, and when sodium selenite at the dose of 5 mumol/kg was given at the time, the effect of cadmium chloride on hepatocellular protooncogene c-myc, c-fos and c-jun expression was inhibited. Cadmium Chloride 33-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 300-305 11769627-6 2001 CONCLUSION: Selenium at certain doses could inhibit hepatocellular protooncogene c-myc, c-fos and c-jun expression induced by cadmium in rats. Selenium 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 11769627-6 2001 CONCLUSION: Selenium at certain doses could inhibit hepatocellular protooncogene c-myc, c-fos and c-jun expression induced by cadmium in rats. Cadmium 126-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 11769628-0 2001 [Effect of methylmercury chloride on the c-fos expression in brain nerve cells of the rats]. Chlorides 25-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 11769628-1 2001 OBJECTIVE: In order to study the mechanism of injury in the brain development caused by methylmercury chloride (MMC) and its effects on the c-fos expression in brain nerve cells of the rats with experiments in vitro and in vivo. methylmercuric chloride 88-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 11769628-1 2001 OBJECTIVE: In order to study the mechanism of injury in the brain development caused by methylmercury chloride (MMC) and its effects on the c-fos expression in brain nerve cells of the rats with experiments in vitro and in vivo. methylmercuric chloride 112-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 11769628-2 2001 METHODS: In vitro, the effect of MMC on c-fos expression in the cultured nerve cells was observed by immunocytochemistry(SP method). methylmercuric chloride 33-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 11769628-2 2001 METHODS: In vitro, the effect of MMC on c-fos expression in the cultured nerve cells was observed by immunocytochemistry(SP method). TFF2 protein, human 121-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 11769628-5 2001 The expression of c-fos was detected by SP method. TFF2 protein, human 40-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 11769628-6 2001 RESULTS: The percentage of c-Fos-positive neuron in rats brain began to increase at 0.5 hours after being cultured in in vitro with 0.3 mumol/L MMC continuously, and increased gradually as length of the exposure to MMC prolonged. methylmercuric chloride 144-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 11769628-6 2001 RESULTS: The percentage of c-Fos-positive neuron in rats brain began to increase at 0.5 hours after being cultured in in vitro with 0.3 mumol/L MMC continuously, and increased gradually as length of the exposure to MMC prolonged. methylmercuric chloride 215-218 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 11769628-7 2001 The percentage of c-Fos-positive of cultured nerve cells in vitro were (6.97 +/- 2.86)%, (66.86 +/- 5.32)% and (64.49 +/- 3.09)% in the experimental groups exposed to MMC for 10 min, 2 h and 6 h, respectively. methylmercuric chloride 167-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 11769628-11 2001 CONCLUSIONS: It is concluded that MMC could induce c-fos over-expression in the nerve cells of the rat brain, which could explain the mechanism of injury to brain development caused by MMC. methylmercuric chloride 34-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 11489249-0 2001 MK-801 reduces non-noxious stimulus-evoked Fos-like immunoreactivity in the spinal cord of rats with chronic constriction nerve injury. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 11489249-1 2001 We investigated the role of N-methyl-D-aspartate (NMDA) receptors on non-noxious stimulus-induced pain by examining the effect of MK-801, a non-competitive NMDA receptor antagonist, on Fos-like immunoreactivity (FLI) in the spinal dorsal horn by non-noxious stimulation to rats with chronic constriction injury (CCI) of the sciatic nerve. Dizocilpine Maleate 130-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 185-188 11478926-8 2001 Addition of atropine to the dialysis medium attenuated the increase of Fos-IR and suppressed the cholinergic stimulation-induced responses in body temperature and water intake. Atropine 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 11447037-4 2001 Hexamethonium (20 mg/kg) also prevented 3-h cold exposure-induced myenteric Fos expression by 76-80%, whereas atropine or bretylium had no effect. Hexamethonium 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 11506121-6 2001 RESULTS: In Fischer rats, 90 min of 75% N2O administration increased the number of c-Fos-positive cells in the spinal cord approximately threefold as compared with the control group. Nitrous Oxide 40-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 11506121-7 2001 The c-Fos-positive cells induced by nitrous oxide were almost entirely colocalized with glutamic acid decarboxylase-positive cells. Nitrous Oxide 36-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 11595358-0 2001 Effects of acute and chronic fluoxetine treatments on restraint stress-induced Fos expression. Fluoxetine 29-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 11490314-13 2001 Pretreatment with MK-801 inhibited bladder overactivity and significantly reduced the expression of c-fos and zif268 mRNA in the pontine tegmental area. Dizocilpine Maleate 18-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 11595358-3 2001 The objective of the present work was to investigate the effects of chronic and acute treatment with fluoxetine (FLX), a selective serotonin reuptake blocker, on Fos expression in animals submitted to restraint stress. Fluoxetine 101-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-165 11595358-3 2001 The objective of the present work was to investigate the effects of chronic and acute treatment with fluoxetine (FLX), a selective serotonin reuptake blocker, on Fos expression in animals submitted to restraint stress. Fluoxetine 113-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-165 11595358-13 2001 The results suggest that chronic fluoxetine treatment induce plastic changes that result in a different regional pattern of Fos expression. Fluoxetine 33-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-127 11553293-6 2001 Nociceptor afferent-induced Fos, detected by immunohistochemistry in superficial and deep dorsal horn laminae, was attenuated by Rp-cAMP, bisindolymaleimide I and GR82334. Cyclic AMP 132-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 11553293-7 2001 In spinal cords pretreated with TTX to eliminate indirect neuronal activation, [Sar9,Met(O2)11]-SP (1-20 microM) elicited a dose-related expression of Fos that was reduced by Rp-cAMP, bisindolymaleimide I and GR82334. Tetrodotoxin 32-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-154 11553293-7 2001 In spinal cords pretreated with TTX to eliminate indirect neuronal activation, [Sar9,Met(O2)11]-SP (1-20 microM) elicited a dose-related expression of Fos that was reduced by Rp-cAMP, bisindolymaleimide I and GR82334. met(o2)11]-sp 85-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-154 11553293-7 2001 In spinal cords pretreated with TTX to eliminate indirect neuronal activation, [Sar9,Met(O2)11]-SP (1-20 microM) elicited a dose-related expression of Fos that was reduced by Rp-cAMP, bisindolymaleimide I and GR82334. Cyclic AMP 178-182 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-154 11553293-7 2001 In spinal cords pretreated with TTX to eliminate indirect neuronal activation, [Sar9,Met(O2)11]-SP (1-20 microM) elicited a dose-related expression of Fos that was reduced by Rp-cAMP, bisindolymaleimide I and GR82334. bisindolymaleimide i 184-204 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-154 11425506-4 2001 Olanzapine prevented induction of striatal Fos protein by SKF-38393 and partially attenuated the long-term "priming" effect of repeated SKF-38393 treatment. Olanzapine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 11454327-10 2001 drastically reduced the induction of Fos-IR neurons in all subnuclei of both the ipsilateral and contralateral PBN in a dose-dependent manner, and its effect was antagonized by pretreatment with naloxone (2 mg/kg, i.p.). Naloxone 195-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 11438395-7 2001 Integrated density of c-fos and PPD mRNA expression was increased in the spinal dorsal horn following QUIS injection as compared to sham-injected animals. Quisqualic Acid 102-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 11454327-11 2001 The reduction of Fos-IR neurons by morphine pretreatment suggests that the appearance of Fos-IR neurons in the PBN may be partly due to the noxious stimulation and/or stress arising from tooth movement. Morphine 35-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 11454327-11 2001 The reduction of Fos-IR neurons by morphine pretreatment suggests that the appearance of Fos-IR neurons in the PBN may be partly due to the noxious stimulation and/or stress arising from tooth movement. Morphine 35-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 11421588-0 2001 Spatiotemporal analysis of Fos expression associated with cocaine- and PTZ-induced seizures in prenatally cocaine-treated rats. Cocaine 58-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 11430892-0 2001 The NMDA receptor antagonist MK-801 reduces Fos-like immunoreactivity within the trigeminocervical complex following superior sagittal sinus stimulation in the cat. Dizocilpine Maleate 29-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 11430892-9 2001 In the group treated with MK-801 the median number of Fos-positive cells was reduced to 40 (30-48; P<0.03, n=7). Dizocilpine Maleate 26-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 11430892-10 2001 The large reduction that was observed in SSS stimulation-evoked Fos protein expression following the administration of MK-801, taken together with electrophysiological data, indicates a role for glutamate in neurotransmission within the trigeminocervical complex. Dizocilpine Maleate 119-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 11430892-10 2001 The large reduction that was observed in SSS stimulation-evoked Fos protein expression following the administration of MK-801, taken together with electrophysiological data, indicates a role for glutamate in neurotransmission within the trigeminocervical complex. Glutamic Acid 195-204 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 11516571-3 2001 Otherwise, the acute c-fos mRNA induction in the striatum was abolished by 74 to 89% upon chronic treatment with either haloperidol or risperidone. Haloperidol 120-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 11516571-3 2001 Otherwise, the acute c-fos mRNA induction in the striatum was abolished by 74 to 89% upon chronic treatment with either haloperidol or risperidone. Risperidone 135-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 11404297-7 2001 Postprandial c-Fos protein expression in the NTS was also significantly decreased after pretreatment with the CCK-A receptor antagonist MK329 after both short- and long-term fasting periods. Devazepide 136-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 11406482-5 2001 The induction of iPLA(2)-mediated CREB phosphorylation was further substantiated by the observations that lysoplasmenylcholine increased both the phosphorylation of CREB and the induction of c-fos expression in the absence and presence of BEL. lysoplasmalogens 106-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 191-196 11421588-0 2001 Spatiotemporal analysis of Fos expression associated with cocaine- and PTZ-induced seizures in prenatally cocaine-treated rats. Pentylenetetrazole 71-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 11421588-0 2001 Spatiotemporal analysis of Fos expression associated with cocaine- and PTZ-induced seizures in prenatally cocaine-treated rats. Cocaine 106-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 11421588-2 2001 In order to determine the locus of enhanced seizure susceptibility in the brains of prenatally cocaine-treated rats, we examined the distribution and density of Fos-immunoreactive cells after cocaine- and PTZ-induced seizures in mature rats. Cocaine 95-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-164 11421588-4 2001 Following cocaine-induced seizures, intense c-fos induction was observed in piriform cortex, amygdala, and hippocampus. Cocaine 10-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 11421588-5 2001 Quantification of the number of Fos-immunoreactive cells in the brains of prenatally cocaine-treated versus prenatally saline-treated rats revealed differences in piriform cortex and amygdala that were indicative of a lower threshold in prenatally cocaine-treated female rats. Cocaine 85-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 11421588-5 2001 Quantification of the number of Fos-immunoreactive cells in the brains of prenatally cocaine-treated versus prenatally saline-treated rats revealed differences in piriform cortex and amygdala that were indicative of a lower threshold in prenatally cocaine-treated female rats. Sodium Chloride 119-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 11421588-7 2001 Only females exhibited changes in the number of Fos-immunoreactive cells as a result of prenatal cocaine treatment. Cocaine 97-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 11421588-10 2001 Examination of the pattern of Fos expression at 15-20 min postseizure revealed that the initial site of c-fos induction associated with PTZ-induced seizures appeared to be the piriform cortex, whereas cocaine-induced seizures induced early expression in both piriform cortex and lateral amygdala. Pentylenetetrazole 136-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 11421588-10 2001 Examination of the pattern of Fos expression at 15-20 min postseizure revealed that the initial site of c-fos induction associated with PTZ-induced seizures appeared to be the piriform cortex, whereas cocaine-induced seizures induced early expression in both piriform cortex and lateral amygdala. Pentylenetetrazole 136-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 11463773-4 2001 Both the suppression of spontaneous baroreceptor reflex and Fos expression in nucleus tractus solitarii neurons elicited by Ang II were discernibly attenuated by pretreatment with or comicroinjection into the bilateral nucleus tractus solitarii of a 15-mer antisense c-fos oligonucleotide that targets against the initiation codon of c-fos mRNA. Oligonucleotides 273-288 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 11442774-0 2001 Effect of nitric oxide synthase inhibition on fos expression in the hypothalamus of female rats following central oxytocin and systemic urethane administration. Urethane 136-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 11531896-4 2001 Intradermal injection of capsaicin in the periorbital area increased c-fos expression in nucleus trigeminalis caudalis; this was significantly potentiated by glyceryl trinitrate. Capsaicin 25-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 11531896-4 2001 Intradermal injection of capsaicin in the periorbital area increased c-fos expression in nucleus trigeminalis caudalis; this was significantly potentiated by glyceryl trinitrate. Nitroglycerin 158-177 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 11442774-11 2001 Overall, lactating rats that received L-NAME and oxytocin had a greater number of cells showing Fos-lir in both the SON and PVN. NG-Nitroarginine Methyl Ester 38-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-99 11442774-12 2001 Conversely, L-NAME administration reduced Fos-lir in the SON and PVN in oxytocin-stimulated nonlactating rats. NG-Nitroarginine Methyl Ester 12-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 11442774-13 2001 In urethane-treated rats, L-NAME administration did not change Fos-lir in lactating rats but reduced Fos-lir in nonlactating rats. NG-Nitroarginine Methyl Ester 26-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-104 11370014-6 2001 First, we examined Fos production by FG-labeled RVLM neurons after 2 hours of hydralazine-induced hypotension (to 73 +/- 2 mm Hg) in conscious rats. Hydralazine 78-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 11294744-0 2001 Injection of muscimol in dorsomedial hypothalamus and stress-induced Fos expression in paraventricular nucleus. Muscimol 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 11474651-7 2001 Phenylephrine (PE)-induced activation of baroreceptors resulted in a significant elevation in the numbers of c-Fos-ir neurons in the NTS of control rats. Phenylephrine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 11474651-7 2001 Phenylephrine (PE)-induced activation of baroreceptors resulted in a significant elevation in the numbers of c-Fos-ir neurons in the NTS of control rats. Phenylephrine 15-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 11474651-9 2001 At both 8 and 16 weeks, STZ-induced diabetic rats had significantly fewer c-Fos-ir neurons in the commissural NTS and in the caudal subpostrernal NTS when compared to the non-diabetic control animals receiving PE. Streptozocin 24-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 11382394-3 2001 S-II stimulation in combination with 7-nitro-indazole at a subeffective dose, 5 mg/kg, synergistically reduced the number of cells expressing c-Fos in response to intraplantar injection of formalin in the superficial regions (laminae I and II) of the L4 and L5 spinal dorsal horn in conscious rats, although each had no significant effect. 7-nitroindazole 37-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-147 11382394-3 2001 S-II stimulation in combination with 7-nitro-indazole at a subeffective dose, 5 mg/kg, synergistically reduced the number of cells expressing c-Fos in response to intraplantar injection of formalin in the superficial regions (laminae I and II) of the L4 and L5 spinal dorsal horn in conscious rats, although each had no significant effect. Formaldehyde 189-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-147 11747755-0 2001 NO mediated increase of Fos protein and NMDA1A R mRNA expression in rat spinal cord during morphine withdrawal. Morphine 91-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 11747755-4 2001 Morphine withdrawal increased the expression of Fos protein, NADPH-d positive, and Fos/NADPH-d double-labeled neurons, and they were observed in all the laminae of the rat spinal cord. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 11747755-4 2001 Morphine withdrawal increased the expression of Fos protein, NADPH-d positive, and Fos/NADPH-d double-labeled neurons, and they were observed in all the laminae of the rat spinal cord. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 11747755-5 2001 Intrathecal injection of nNOS antisense oligonucleotides (nNOS-AS) inhibited the increase of Fos protein and NMDA(1A)R mRNA expression in the rat spinal cord during morphine withdrawal and decreased the scores of morphine withdrawal symptoms. Oligonucleotides 40-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 11747755-8 2001 CONCLUSION: NO mediated the increase of Fos protein and NMDA1A R mRNA expression in the rat spinal cord during morphine withdrawal. Morphine 111-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 11353692-3 2001 To test this hypothesis, we determined the effect of deoxycorticosterone (DOCA; 2 mg/day) on icv ANG II-induced c-Fos immunoreactivity in OT and vasopressin (VP) neurons in the supraoptic (SON) and paraventricular (PVN) nuclei of the hypothalamus and also on pituitary OT and VP secretion in male rats. Desoxycorticosterone 53-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 11353692-3 2001 To test this hypothesis, we determined the effect of deoxycorticosterone (DOCA; 2 mg/day) on icv ANG II-induced c-Fos immunoreactivity in OT and vasopressin (VP) neurons in the supraoptic (SON) and paraventricular (PVN) nuclei of the hypothalamus and also on pituitary OT and VP secretion in male rats. Desoxycorticosterone Acetate 74-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 11353692-4 2001 DOCA significantly decreased the percentage of c-Fos-positive (%c-Fos+) OT neurons in the SON and PVN, both in the magnocellular and parvocellular subdivisions, and the %c-Fos+ VP neurons in the SON after a 5-ng icv injection of ANG II. Desoxycorticosterone Acetate 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 11353692-4 2001 DOCA significantly decreased the percentage of c-Fos-positive (%c-Fos+) OT neurons in the SON and PVN, both in the magnocellular and parvocellular subdivisions, and the %c-Fos+ VP neurons in the SON after a 5-ng icv injection of ANG II. Desoxycorticosterone Acetate 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 11353692-4 2001 DOCA significantly decreased the percentage of c-Fos-positive (%c-Fos+) OT neurons in the SON and PVN, both in the magnocellular and parvocellular subdivisions, and the %c-Fos+ VP neurons in the SON after a 5-ng icv injection of ANG II. Desoxycorticosterone Acetate 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 11353692-5 2001 DOCA also significantly reduced the %c-Fos+ OT neurons in the SON after 10 ng ANG II and tended to attenuate 10 ng ANG II-induced OT secretion. Desoxycorticosterone Acetate 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 11275291-0 2001 Nicotine-induced behavioral sensitization is associated with extracellular dopamine release and expression of c-Fos in the striatum and nucleus accumbens of the rat. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 11275291-2 2001 This study was carried out to investigate the neural mechanisms underlying nicotine-induced behavioral sensitization using in vivo microdialysis and Fos-like immunohistochemistry (FLI). Nicotine 75-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 149-152 11275291-6 2001 Furthermore, in parallel with the behavioral and biochemical data, systemic challenge with nicotine produced marked Fos-like immunohistochemistry in the nucleus accumbens and the striatum in the nicotine-pretreated rats. Nicotine 91-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 11275291-6 2001 Furthermore, in parallel with the behavioral and biochemical data, systemic challenge with nicotine produced marked Fos-like immunohistochemistry in the nucleus accumbens and the striatum in the nicotine-pretreated rats. Nicotine 195-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 11275291-7 2001 Taken together, this study demonstrates that behavioral sensitization is clearly associated with an increase in DA release and activation of Fos-like immunoreactive cells in the striatum and the nucleus accumbens produced by repeated nicotine treatment. Nicotine 234-242 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 11337201-4 2001 In situ hybridization histochemistry experiment was performed to study its effects on the induction of immediate early gene expression (c-fos, c-jun) by dopamine D1 receptor agonist SKF-82958 and on the augmentation of the SKF-82958-induced expression of these genes by scopolamine. Scopolamine 270-281 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 11337201-6 2001 We found that physostigmine but not PBN significantly reversed the cognitive impairment in scopolamine-challenged rats, prevented the induction of c-fos and c-jun mRNAs by SKF-82958 and attenuated the augmentation of the SKF-82958-induced expression of these genes by scopolamine. Physostigmine 14-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-152 11356898-6 2001 AP-5 treatment also disrupted mating-induced c-fos expression in the principle bed nucleus of the stria terminalis and the ventrolateral division of the ventromedial hypothalamic nucleus, but not in the medial or anteroventral periventricular preoptic nuclei. 2-amino-5-phosphopentanoic acid 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 11389180-0 2001 The role of putative intragenic control elements in c-fos regulation by calcium and growth factor signalling pathways. Calcium 72-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 11389180-1 2001 Sequences in the transcribed region of the c-fos gene have been suggested to control c-fos induction following exposure of cells to mitogens or stimuli that increase intracellular calcium concentrations. Calcium 180-187 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 11389180-1 2001 Sequences in the transcribed region of the c-fos gene have been suggested to control c-fos induction following exposure of cells to mitogens or stimuli that increase intracellular calcium concentrations. Calcium 180-187 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 11389180-3 2001 Removal of the c-fos first intron and the FIRE did not increase the basal level of c-fos mRNA and only moderately reduced the magnitude of calcium-induced transcription mediated by either the entire c-fos promoter or the cAMP response element (CRE). Calcium 139-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 11389180-3 2001 Removal of the c-fos first intron and the FIRE did not increase the basal level of c-fos mRNA and only moderately reduced the magnitude of calcium-induced transcription mediated by either the entire c-fos promoter or the cAMP response element (CRE). Cyclic AMP 221-225 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 11356297-0 2001 Induction of unspliced c-fos messenger RNA in rodent brain by kainic acid and lipopolysaccharide. Kainic Acid 62-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 11408778-9 2001 Numbers of TH-positive A2 and A6 neurons that expressed Fos in response to 2DG were significantly greater in rats implanted with the high E dose vs. either the low steroid dose or sesame oil. Deoxyglucose 75-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 11408780-2 2001 In order to gain insight into the molecular mechanisms involved, we studied induction of c-fos, an index of functional neuronal activation, in the 2-day-old female rat brain after injection of a masculinizing dose of testosterone. Testosterone 217-229 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 11408780-3 2001 Administration of testosterone resulted in induction of c-fos gene expression in the hypothalamus, as determined by Northern analysis. Testosterone 18-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 11408780-7 2001 Furthermore, injection of an estrogen receptor blocker, clomiphene, together with testosterone, abolished the testosterone-induced increase in Fos-positive nuclei, thus confirming the finding that testosterone induces c-fos by acting through estrogen receptors. Clomiphene 56-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-146 11408780-7 2001 Furthermore, injection of an estrogen receptor blocker, clomiphene, together with testosterone, abolished the testosterone-induced increase in Fos-positive nuclei, thus confirming the finding that testosterone induces c-fos by acting through estrogen receptors. Clomiphene 56-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 218-223 11408780-7 2001 Furthermore, injection of an estrogen receptor blocker, clomiphene, together with testosterone, abolished the testosterone-induced increase in Fos-positive nuclei, thus confirming the finding that testosterone induces c-fos by acting through estrogen receptors. Testosterone 82-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-146 11408780-7 2001 Furthermore, injection of an estrogen receptor blocker, clomiphene, together with testosterone, abolished the testosterone-induced increase in Fos-positive nuclei, thus confirming the finding that testosterone induces c-fos by acting through estrogen receptors. Testosterone 82-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 218-223 11408780-7 2001 Furthermore, injection of an estrogen receptor blocker, clomiphene, together with testosterone, abolished the testosterone-induced increase in Fos-positive nuclei, thus confirming the finding that testosterone induces c-fos by acting through estrogen receptors. Testosterone 110-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-146 11408780-7 2001 Furthermore, injection of an estrogen receptor blocker, clomiphene, together with testosterone, abolished the testosterone-induced increase in Fos-positive nuclei, thus confirming the finding that testosterone induces c-fos by acting through estrogen receptors. Testosterone 110-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 218-223 11408780-7 2001 Furthermore, injection of an estrogen receptor blocker, clomiphene, together with testosterone, abolished the testosterone-induced increase in Fos-positive nuclei, thus confirming the finding that testosterone induces c-fos by acting through estrogen receptors. Testosterone 110-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-146 11408780-7 2001 Furthermore, injection of an estrogen receptor blocker, clomiphene, together with testosterone, abolished the testosterone-induced increase in Fos-positive nuclei, thus confirming the finding that testosterone induces c-fos by acting through estrogen receptors. Testosterone 110-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 218-223 11408780-9 2001 This suggests that the effect of testosterone-derived estradiol on c-fos expression is a direct one, mediated by binding of estrogen receptors to an ERE in the c-fos gene-regulatory regions. Testosterone 33-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 11408780-9 2001 This suggests that the effect of testosterone-derived estradiol on c-fos expression is a direct one, mediated by binding of estrogen receptors to an ERE in the c-fos gene-regulatory regions. Testosterone 33-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 11376912-6 2001 morphine reduced by 30% the number of Fos-LI neurones induced by heat stimulation (52 degrees C, 15 s duration) in CCI rats (P < 0.05) as in sham-operated rats. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 11484778-7 2001 Addition of atropine, a muscarinic receptor antagonist, to the dialysis medium containing neostigmine attenuated the increase of Fos-IR and suppressed the neostigmine-induced responses in body temperature. Atropine 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-132 11484778-7 2001 Addition of atropine, a muscarinic receptor antagonist, to the dialysis medium containing neostigmine attenuated the increase of Fos-IR and suppressed the neostigmine-induced responses in body temperature. Neostigmine 90-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-132 11465628-0 2001 Cross tolerance between anorectic action and induction of Fos-ir with dexfenfluramine and 5HT1B/2C agonists in rats. Dexfenfluramine 70-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 11465628-11 2001 Fos-ir induced by DFEN in each brain region examined was either significantly reduced or abolished by prior DFEN injections. Dexfenfluramine 18-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 11465628-11 2001 Fos-ir induced by DFEN in each brain region examined was either significantly reduced or abolished by prior DFEN injections. Dexfenfluramine 108-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 11465628-12 2001 TFMPP induced less Fos-ir in these regions than DFEN and this was attenuated by prior DFEN. 1-(3-trifluoromethylphenyl)piperazine 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 11376854-10 2001 During chronic osmotic stimulation by giving 2% NaCl solution for 2 and 5 days, a large number of Fos-positive neurons were observed, but the cessation of chronic osmotic stimulation by normal water drinking immediately decreased the number of Fos-positive neurons to the control level within 2 h. The number of FosB-positive neurons was increased with period of chronic osmotic stimulation, and a significant number were observed 2-8 h after the cessation of the stimulation. nacl solution 48-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 11376854-10 2001 During chronic osmotic stimulation by giving 2% NaCl solution for 2 and 5 days, a large number of Fos-positive neurons were observed, but the cessation of chronic osmotic stimulation by normal water drinking immediately decreased the number of Fos-positive neurons to the control level within 2 h. The number of FosB-positive neurons was increased with period of chronic osmotic stimulation, and a significant number were observed 2-8 h after the cessation of the stimulation. nacl solution 48-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 244-247 11376854-10 2001 During chronic osmotic stimulation by giving 2% NaCl solution for 2 and 5 days, a large number of Fos-positive neurons were observed, but the cessation of chronic osmotic stimulation by normal water drinking immediately decreased the number of Fos-positive neurons to the control level within 2 h. The number of FosB-positive neurons was increased with period of chronic osmotic stimulation, and a significant number were observed 2-8 h after the cessation of the stimulation. Water 193-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 244-247 11340648-0 2001 Inhibitory effect of neuropeptide Y on morphine withdrawal is accompanied by reduced c-fos expression in specific brain regions. Morphine 39-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 11340648-6 2001 Brain areas involved in the attenuation of morphine withdrawal were delineated by radioactive in situ hybridization for the immediate early gene c-fos, which is a marker for neuronal activity. Morphine 43-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-150 11340648-8 2001 Inhibition of behavioral signs of naloxone-precipitated morphine withdrawal was accompanied by significantly reduced c-fos expression in the locus coeruleus, lateral septal nucleus, ventral part of the periaqueductal grey, cingulate and frontal cortices, and septohippocampal nucleus. Naloxone 34-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 11340648-8 2001 Inhibition of behavioral signs of naloxone-precipitated morphine withdrawal was accompanied by significantly reduced c-fos expression in the locus coeruleus, lateral septal nucleus, ventral part of the periaqueductal grey, cingulate and frontal cortices, and septohippocampal nucleus. Morphine 56-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 11311502-0 2001 Nomifensine-induced c-fos mRNA expression in discrete brain areas of the developing rat. Nomifensine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 11311502-1 2001 In rats, the subcutaneous injection of a dopamine uptake inhibitor, nomifensine (40 mg/kg), induced a significant increase in the c-fos mRNA levels in the neocortex on postnatal days 23 and 49, in the striatum on days 8, 14, 23 and 49, and in the hippocampus on day 23, when compared with saline administration. Dopamine 41-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 11311502-1 2001 In rats, the subcutaneous injection of a dopamine uptake inhibitor, nomifensine (40 mg/kg), induced a significant increase in the c-fos mRNA levels in the neocortex on postnatal days 23 and 49, in the striatum on days 8, 14, 23 and 49, and in the hippocampus on day 23, when compared with saline administration. Nomifensine 68-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 11371719-0 2001 Expression of c-Fos in Alko alcohol rats responding for ethanol in an operant paradigm. Alcohols 28-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 11371719-0 2001 Expression of c-Fos in Alko alcohol rats responding for ethanol in an operant paradigm. Ethanol 56-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 11371719-3 2001 As an extension of studies mapping changes in neural activity after voluntary ethanol drinking, this study analyzed expression of the inducible transcription factor c-Fos after ethanol consumption in an operant procedure. Ethanol 78-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 11371719-3 2001 As an extension of studies mapping changes in neural activity after voluntary ethanol drinking, this study analyzed expression of the inducible transcription factor c-Fos after ethanol consumption in an operant procedure. Ethanol 177-184 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 11371719-7 2001 RESULTS: In this paradigm, ethanol dose-dependently increased c-Fos expression in the Edinger-Westphal nucleus (EW) and decreased expression in the dorsal tenia tecta compared with no-ethanol controls. Ethanol 27-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 11371719-10 2001 The finding that ethanol attenuated c-Fos expression in the tenia tecta is novel. Ethanol 17-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 11294744-4 2001 Each stressor evoked a characteristic pattern of Fos expression in the parvocellular and magnocellular PVN after saline. Sodium Chloride 113-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 11294744-5 2001 Injection of muscimol into the DMH suppressed Fos expression in the PVN associated with air stress but not with hemorrhage. Muscimol 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 11294744-5 2001 Injection of muscimol into the DMH suppressed Fos expression in the PVN associated with air stress but not with hemorrhage. 1,2-Dimethylhydrazine 31-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 11325566-0 2001 Time-dependent distribution and neuronal localization of c-fos protein in the rat hippocampus following 4-aminopyridine seizures. 4-Aminopyridine 104-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 11292608-4 2001 Hexamethonium (20 mg/kg sc) prevented Fos expression by 90%, whereas atropine (2 mg/kg sc) had no effect. Hexamethonium 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 11401756-2 2001 Adenosine-5"-triphosphate (ATP) was determined to increase and, at higher concentrations, decrease luciferase activity in GH(4)C(1) rat pituitary cells that stably express c-fos luciferase. Adenosine Triphosphate 0-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-177 11401756-2 2001 Adenosine-5"-triphosphate (ATP) was determined to increase and, at higher concentrations, decrease luciferase activity in GH(4)C(1) rat pituitary cells that stably express c-fos luciferase. Adenosine Triphosphate 27-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-177 11401756-4 2001 The actions of both pPfTx and ATP to induce c-fos luciferase were inhibited by the purinogenic receptor antagonist pyridoxalphosphate-6-azophenyl-2",4"-disulfonic acid (PPADS). ppftx 20-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 11401756-4 2001 The actions of both pPfTx and ATP to induce c-fos luciferase were inhibited by the purinogenic receptor antagonist pyridoxalphosphate-6-azophenyl-2",4"-disulfonic acid (PPADS). Adenosine Triphosphate 30-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 11401756-4 2001 The actions of both pPfTx and ATP to induce c-fos luciferase were inhibited by the purinogenic receptor antagonist pyridoxalphosphate-6-azophenyl-2",4"-disulfonic acid (PPADS). pyridoxal phosphate-6-azophenyl-2',4'-disulfonic acid 115-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 11401756-10 2001 These findings indicate that GH(4)C(1) cells express purinogenic receptors with selectivity consistent with the P2X7 subtype and that this receptor pathway mediates the induction of the c-fos luciferase reporter gene by ATP and the putative Pfiesteria toxin Adenosine Triphosphate 220-223 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 186-191 11325566-1 2001 The immunohistochemical localization of c-fos protein in the CNS neurons was studied in a model of generalized epilepsy induced by the intraperitoneal injection of 4-aminopyridine to adult Wistar rats. 4-Aminopyridine 164-179 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 11267996-5 2001 ATP, UTP, UDP and ATPgammaS increased (3)H-thymidine incorporation into DNA and expression of the protooncogenes c-Fos and c-Jun, while 2-MeSATP was ineffective, and alpha,beta-meATP gave a response only at 100-microM dose. Adenosine Triphosphate 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 11333365-5 2001 Administration of N-acetyl-O-methyldopamine (NAMDA), a compound previously shown to protect CA1 neurons against ischemia, increased c-Fos immunoreactivity in the CA1 vulnerable region at 6 hours after ischemia and protected SK-N-BE(2)C neurons from oxygen glucose deprivation. Acetamide, N-[2-(4-hydroxy-3-methoxyphenyl)ethyl]- 18-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 11333365-5 2001 Administration of N-acetyl-O-methyldopamine (NAMDA), a compound previously shown to protect CA1 neurons against ischemia, increased c-Fos immunoreactivity in the CA1 vulnerable region at 6 hours after ischemia and protected SK-N-BE(2)C neurons from oxygen glucose deprivation. namda 45-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 11333365-5 2001 Administration of N-acetyl-O-methyldopamine (NAMDA), a compound previously shown to protect CA1 neurons against ischemia, increased c-Fos immunoreactivity in the CA1 vulnerable region at 6 hours after ischemia and protected SK-N-BE(2)C neurons from oxygen glucose deprivation. sk-n-be 224-231 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 11333365-6 2001 Further in vitro study showed that NAMDA potentiated phorbol-12 myristate-13 acetate (PMA)-induced c-Fos expression, AP1 binding activity, and late gene expression determined by chloramphenicol acetyltransferase (CAT) activity from AP1 containing tyrosine hydroxylase promoter-CAT fusion gene in SK-N-BE(2)C neurons. namda 35-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 11333365-6 2001 Further in vitro study showed that NAMDA potentiated phorbol-12 myristate-13 acetate (PMA)-induced c-Fos expression, AP1 binding activity, and late gene expression determined by chloramphenicol acetyltransferase (CAT) activity from AP1 containing tyrosine hydroxylase promoter-CAT fusion gene in SK-N-BE(2)C neurons. Tetradecanoylphorbol Acetate 53-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 11333365-6 2001 Further in vitro study showed that NAMDA potentiated phorbol-12 myristate-13 acetate (PMA)-induced c-Fos expression, AP1 binding activity, and late gene expression determined by chloramphenicol acetyltransferase (CAT) activity from AP1 containing tyrosine hydroxylase promoter-CAT fusion gene in SK-N-BE(2)C neurons. Tetradecanoylphorbol Acetate 86-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 11333365-7 2001 In vivo and in vitro results showed that a neuroprotectant, NAMDA, in concert with another stimulus (for example, ischemia or PMA) up-regulates c-Fos expression and suggested that the early rise of NAMDA-induced c-Fos expression in vulnerable CA1 neurons may account for neuroprotection by means of up-regulating late gene expression for survival. namda 60-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-149 11333365-7 2001 In vivo and in vitro results showed that a neuroprotectant, NAMDA, in concert with another stimulus (for example, ischemia or PMA) up-regulates c-Fos expression and suggested that the early rise of NAMDA-induced c-Fos expression in vulnerable CA1 neurons may account for neuroprotection by means of up-regulating late gene expression for survival. namda 60-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 212-217 11333365-7 2001 In vivo and in vitro results showed that a neuroprotectant, NAMDA, in concert with another stimulus (for example, ischemia or PMA) up-regulates c-Fos expression and suggested that the early rise of NAMDA-induced c-Fos expression in vulnerable CA1 neurons may account for neuroprotection by means of up-regulating late gene expression for survival. namda 198-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-149 11333365-7 2001 In vivo and in vitro results showed that a neuroprotectant, NAMDA, in concert with another stimulus (for example, ischemia or PMA) up-regulates c-Fos expression and suggested that the early rise of NAMDA-induced c-Fos expression in vulnerable CA1 neurons may account for neuroprotection by means of up-regulating late gene expression for survival. namda 198-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 212-217 11267996-5 2001 ATP, UTP, UDP and ATPgammaS increased (3)H-thymidine incorporation into DNA and expression of the protooncogenes c-Fos and c-Jun, while 2-MeSATP was ineffective, and alpha,beta-meATP gave a response only at 100-microM dose. Uridine Triphosphate 5-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 11267996-5 2001 ATP, UTP, UDP and ATPgammaS increased (3)H-thymidine incorporation into DNA and expression of the protooncogenes c-Fos and c-Jun, while 2-MeSATP was ineffective, and alpha,beta-meATP gave a response only at 100-microM dose. Uridine Diphosphate 10-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 11267996-5 2001 ATP, UTP, UDP and ATPgammaS increased (3)H-thymidine incorporation into DNA and expression of the protooncogenes c-Fos and c-Jun, while 2-MeSATP was ineffective, and alpha,beta-meATP gave a response only at 100-microM dose. adenosine 5'-O-(3-thiotriphosphate) 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 11267996-6 2001 The ATP-stimulated expression of c-Fos and c-Jun was dependent on Ca(2+), and protein kinase C, but not on calmodulin or Ca(2+)/calmodulin-dependent protein kinase II. Adenosine Triphosphate 4-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 11267996-7 2001 Extracellular signal-regulated kinases (ERK1 and ERK2) are also involved as the MEK inhibitor, PD98059, reduced both ATP-evoked (3)H-thymidine incorporation and c-Fos and c-Jun expression. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 95-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 11378256-0 2001 The acetylcholine release enhancer linopirdine induces Fos in neocortex of aged rats. Acetylcholine 4-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 11378256-0 2001 The acetylcholine release enhancer linopirdine induces Fos in neocortex of aged rats. linopirdine 35-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 11378256-2 2001 In this study we have monitored by Fos immunohistochemistry the effect of the acetylcholine release enhancer linopirdine (DUP996) on the immediate early gene c-fos in brains of 3 months and 30 months old rats. Acetylcholine 78-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 11378256-2 2001 In this study we have monitored by Fos immunohistochemistry the effect of the acetylcholine release enhancer linopirdine (DUP996) on the immediate early gene c-fos in brains of 3 months and 30 months old rats. linopirdine 109-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 11378256-2 2001 In this study we have monitored by Fos immunohistochemistry the effect of the acetylcholine release enhancer linopirdine (DUP996) on the immediate early gene c-fos in brains of 3 months and 30 months old rats. linopirdine 109-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 11378256-3 2001 In young rats linopirdine had only a marginal effect on Fos expression. linopirdine 14-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 11378256-4 2001 In contrast, in aged rats linopirdine caused widespread expression of Fos throughout neocortex. linopirdine 26-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 11378256-5 2001 In somatosensory cortex, the induction of the c-fos gene by linopirdine was nearly completely blocked by atropine and scopolamine and strongly attenuated by the NMDA receptor blockers CPP and MK-801. linopirdine 60-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 11378256-5 2001 In somatosensory cortex, the induction of the c-fos gene by linopirdine was nearly completely blocked by atropine and scopolamine and strongly attenuated by the NMDA receptor blockers CPP and MK-801. Atropine 105-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 11378256-5 2001 In somatosensory cortex, the induction of the c-fos gene by linopirdine was nearly completely blocked by atropine and scopolamine and strongly attenuated by the NMDA receptor blockers CPP and MK-801. Scopolamine 118-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 11378256-5 2001 In somatosensory cortex, the induction of the c-fos gene by linopirdine was nearly completely blocked by atropine and scopolamine and strongly attenuated by the NMDA receptor blockers CPP and MK-801. Dizocilpine Maleate 192-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 11399902-8 2001 Pretreatment with a selective CRH-R1 antagonist, CP-154,526, significantly attenuated stress-induced corticotropin (ACTH) secretion as well as c-fos mRNA expression in the PVN. cp-154 49-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-148 11323123-6 2001 Hypertonic saline (0.5 M) caused only a minor expression of c-fos mRNA in the hypothalamus and amygdala. Sodium Chloride 0-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 11283964-1 2001 Several studies have used c-Fos expression to delineate the neural substrate underlying naloxone-precipitated morphine withdrawal (MW). Naloxone 88-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 11283964-1 2001 Several studies have used c-Fos expression to delineate the neural substrate underlying naloxone-precipitated morphine withdrawal (MW). Morphine 110-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 11355693-1 2001 Mitogen-activated protein kinases (MAPKs) may play crucial roles in the kainic acid (KA)-evoked excitotoxic effect and the regulation of transcription factors (e.g. c-Fos and c-Jun) in hippocampus, but their exact role in the regulation of KA-induced opioid peptides expression has not been well characterized in vivo. Kainic Acid 72-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 11297813-0 2001 The neurotensin antagonist SR 48692 attenuates haloperidol-induced striatal Fos expression in the rat. SR 48692 27-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 11297813-0 2001 The neurotensin antagonist SR 48692 attenuates haloperidol-induced striatal Fos expression in the rat. Haloperidol 47-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 11297813-5 2001 SR 48692 reduced Fos expression in the striatal patch (striosome) and matrix compartments, with a significantly greater effect in the patch. SR 48692 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 11290403-3 2001 In this work we use c-fos-like immunoreactivity to detect active areas involved in the long-term control of increased water and sodium intake due to partial aortic ligature. Water 118-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 11290403-3 2001 In this work we use c-fos-like immunoreactivity to detect active areas involved in the long-term control of increased water and sodium intake due to partial aortic ligature. Sodium 128-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 11303770-6 2001 The number of nitric oxide-evoked Fos-immunoreactive cells between the two groups remained comparable. Nitric Oxide 14-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 11334850-0 2001 M100907, a selective 5-HT(2A) receptor antagonist, attenuates phencyclidine-induced Fos expression in discrete regions of rat brain. volinanserin 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 11334850-0 2001 M100907, a selective 5-HT(2A) receptor antagonist, attenuates phencyclidine-induced Fos expression in discrete regions of rat brain. Phencyclidine 62-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 11334850-4 2001 For the purpose of identifying regions in which M100907 exerts its effect, we investigated the effects of M100907 on PCP-induced Fos expression in rat brain. Phencyclidine 117-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-132 11334850-5 2001 PCP (5 mg/kg, subcutaneously, s.c.) induced Fos expression in the cingulate cortex area 3, the agranular insular cortex, the piriform cortex, the nucleus accumbens, the anterior paraventricular thalamic nucleus and the ventral lateral septal nucleus. Phencyclidine 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 11334850-6 2001 Pretreatment with M100907 (0.5 mg/kg, s.c.) attenuated Fos expression induced by PCP in the nucleus accumbens core, the shell, the agranular insular cortex and the piriform cortex. Phencyclidine 81-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 11334850-8 2001 These results indicate that 5-HT(2A) receptor antagonism attenuates Fos expression in a regionally specific manner in rat brain in the PCP model of psychosis. Phencyclidine 135-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 11282356-5 2001 Administration of the NMDA receptor antagonist MK-801 (3 mg/kg) prior to light pulses had no effect on c-fos in the first part of the night, but towards the expected time of lights on, became progressively more potent, such that by CT0, light induction of c-fos was almost completely inhibited. Dizocilpine Maleate 47-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 256-261 11282380-1 2001 Three priming injections with the D1/D2 dopamine agonist apomorphine permits a challenge with the D2 agonist quinpirole to elicit robust contralateral rotation and ipsilateral striatal Fos expression in 6-hydroxydopamine lesioned rats. Dopamine 40-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 185-188 11282380-1 2001 Three priming injections with the D1/D2 dopamine agonist apomorphine permits a challenge with the D2 agonist quinpirole to elicit robust contralateral rotation and ipsilateral striatal Fos expression in 6-hydroxydopamine lesioned rats. Apomorphine 57-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 185-188 11282380-1 2001 Three priming injections with the D1/D2 dopamine agonist apomorphine permits a challenge with the D2 agonist quinpirole to elicit robust contralateral rotation and ipsilateral striatal Fos expression in 6-hydroxydopamine lesioned rats. Quinpirole 109-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 185-188 11282380-1 2001 Three priming injections with the D1/D2 dopamine agonist apomorphine permits a challenge with the D2 agonist quinpirole to elicit robust contralateral rotation and ipsilateral striatal Fos expression in 6-hydroxydopamine lesioned rats. Oxidopamine 203-220 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 185-188 11301070-0 2001 Clozapine-induced Fos-protein expression in rat forebrain regions: differential effects of adrenalectomy and corticosterone supplement. Clozapine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 11301070-1 2001 Unlike classical antipsychotic drugs, clozapine activates the hypothalamo-pituitary-adrenal axis and induces a specific regional pattern of Fos-protein expression in the rat forebrain. Clozapine 38-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-143 11301070-2 2001 Whether corticosterone plays a role in the clozapine-induced Fos response is the subject of this study. Corticosterone 8-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 11301070-2 2001 Whether corticosterone plays a role in the clozapine-induced Fos response is the subject of this study. Clozapine 43-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 11301070-5 2001 The clozapine-induced Fos response was not affected by adrenalectomy, apart from the nucleus accumbens shell, the subfornical organ and the supraoptic nucleus; there was an increased response in the nucleus accumbens shell, while other regions showed less Fos immunoreactivity. Clozapine 4-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-25 11301070-5 2001 The clozapine-induced Fos response was not affected by adrenalectomy, apart from the nucleus accumbens shell, the subfornical organ and the supraoptic nucleus; there was an increased response in the nucleus accumbens shell, while other regions showed less Fos immunoreactivity. Clozapine 4-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 256-259 11301070-6 2001 Implantation of the corticosterone pellet in both sham-operated and adrenalectomized animals, reduced the clozapine-induced Fos responses strongly in the hypothalamic paraventricular nucleus, the subfornical organ and possibly in the prefrontal cortex; in the supraoptic nucleus, this effect was seen only in intact animals. Corticosterone 20-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-127 11301070-6 2001 Implantation of the corticosterone pellet in both sham-operated and adrenalectomized animals, reduced the clozapine-induced Fos responses strongly in the hypothalamic paraventricular nucleus, the subfornical organ and possibly in the prefrontal cortex; in the supraoptic nucleus, this effect was seen only in intact animals. Clozapine 106-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-127 11241386-4 2001 2DG-induced Fos expression was profoundly reduced or abolished in the PVH, but not in the adrenal medulla. Deoxyglucose 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-15 11241386-7 2001 2DG-induced Fos-ir was abolished in the adrenal medulla but not in the PVH. Deoxyglucose 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-15 11279288-4 2001 Eticlopride-induced c-fos and zif268 mRNA expression in striatum was not blocked by H-89. eticlopride 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 11306007-3 2001 In the SON and anterior pituitary, a large number of Fos-positive cells were observed by restraint stress, hyperosmotic administration (1.5, 3, and 9% NaCl), and LPS administration (5, 25, and 125 microg/kg). Sodium Chloride 151-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 11259263-1 2001 A hallmark of reproductive aging in rats is a delay in the initiation and peak, and a decrease in the amplitude, of both proestrous and steroid-induced surges of LH and a decrease in the number of GnRH neurons that express Fos during the surge. Steroids 136-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 223-226 11599124-4 2001 Heparin inhibited ET-1-induced c-fos mRNA expression. Heparin 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 11599124-5 2001 Heparin and HS inhibited ET-1-induced activation of c-fos promoter/enhancer in MCs. Heparin 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 11599124-6 2001 Although heparin and HS inhibited ET-1-induced activation of the wild-type c-fos serum response element (SRE), the activation of a mutated c-fos SRE that contains an intact binding site for the serum response factor (SRF) but lacks the ternary complex factor (TCF) binding site, was not inhibited. Heparin 9-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 11599124-10 2001 These results indicate that heparin and HS inhibited ET-1-induced ERK activation, resulting in suppression of Elk-1 phosphorylation, and lead to inhibition of c-fos gene expression through SRF-independent manner. Heparin 28-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-164 11471222-4 2001 Intracerebroventricular injection of atropine partly blocked the SFO stimulation-induced drinking behavior and the Fos protein expression in the brain, suggesting that an M-cholinergic mechanism may be involved. Atropine 37-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-118 11170217-0 2001 Neonatal phencyclidine treatment selectively attenuates mesolimbic dopamine function in adult rats as revealed by methamphetamine-induced behavior and c-fos mRNA expression in the brain. Phencyclidine 9-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-156 11248440-1 2001 Here we examine hypothesis that short-term peripheral ZnSO(4)-induced anosmia can produce effects on c-fos expression within spinal cord and caudal medulla in male Wistar rats (n=4). Zinc Sulfate 54-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 11124938-12 2001 In fact the calcium responsive Ca/cAMP response element of the c-fos promoter alone was effective at driving this synergistic gene expression. Calcium 12-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 11124938-12 2001 In fact the calcium responsive Ca/cAMP response element of the c-fos promoter alone was effective at driving this synergistic gene expression. Cyclic AMP 34-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 11251193-11 2001 MK-801 blocked these changes in ITF expressions, but it could also cause the C-fibre stimulations to induce c-Fos and c-Jun in specific areas of the hippocampus. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 11223008-0 2001 Comparative effects of scopolamine and quinpirole on the striatal fos expression induced by stimulation of D(1) dopamine receptors in the rat. Scopolamine 23-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 11223008-0 2001 Comparative effects of scopolamine and quinpirole on the striatal fos expression induced by stimulation of D(1) dopamine receptors in the rat. Quinpirole 39-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 11223008-1 2001 Treatment of intact rats with the full D(1) dopamine agonist A-77636 induced Fos-like immunoreactivity in the medial and, to a lesser extent, the lateral portions of the striatum. d(1) dopamine 39-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-80 11223008-7 2001 These findings demonstrate that there are both differences and similarities between the effects of scopolamine and quinpirole on D(1) agonist-induced Fos expression and suggest that although inhibition of cholinergic neurons may be one of the mechanisms through which the effects of quinpirole are produced, other factors must also contribute. Scopolamine 99-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-153 11223008-7 2001 These findings demonstrate that there are both differences and similarities between the effects of scopolamine and quinpirole on D(1) agonist-induced Fos expression and suggest that although inhibition of cholinergic neurons may be one of the mechanisms through which the effects of quinpirole are produced, other factors must also contribute. Quinpirole 115-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-153 11171624-11 2001 Capsaicin treatment not only significantly reduced the Fos-positive neuron numbers in portal hypertensive and cirrhotic rats but also attenuated hemorrhage-induced Fos and double-positive cells in both NTS and VLM. Capsaicin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 11171624-11 2001 Capsaicin treatment not only significantly reduced the Fos-positive neuron numbers in portal hypertensive and cirrhotic rats but also attenuated hemorrhage-induced Fos and double-positive cells in both NTS and VLM. Capsaicin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-167 11357518-0 2001 Increased expression of iNOS and c-fos via regulation of protein tyrosine phosphorylation and MEK1/ERK2 proteins in terminal bronchiole lesions in the lungs of rats exposed to cigarette smoke. Tyrosine 65-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 11244024-3 2001 Previously we showed that pressure-induced c-fos expression in intact cannulated rat mesenteric small arteries was inhibited by genistein, a general tyrosine kinase inhibitor. Genistein 128-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 11327629-4 2001 c-fos mRNA expression by angiotensin II was determined by reverse transcriptase-polymerase chain reaction in the absence and presence of PD98059, selective inhibitor of ERK1/2-dependent pathways and SB202190, selective p38MAPK inhibitor. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 137-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 11327629-4 2001 c-fos mRNA expression by angiotensin II was determined by reverse transcriptase-polymerase chain reaction in the absence and presence of PD98059, selective inhibitor of ERK1/2-dependent pathways and SB202190, selective p38MAPK inhibitor. 4-(4-fluorophenyl)-2-(4-hydroxyphenyl)-5-(4-pyridyl)imidazole 199-207 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 11222656-7 2001 Sleep deprivation or treatment with methamphetamine also increased Fos expression in orexin neurons. Methamphetamine 36-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 11166331-5 2001 Double activity imaging revealed that haloperidol, clozapine and RGH-1756 share cellular targets in the nucleus accumbens, where 40% of all labeled neurons displayed both c-fos mRNA and c-Fos protein. Haloperidol 38-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-176 11166331-5 2001 Double activity imaging revealed that haloperidol, clozapine and RGH-1756 share cellular targets in the nucleus accumbens, where 40% of all labeled neurons displayed both c-fos mRNA and c-Fos protein. Haloperidol 38-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 186-191 11166331-5 2001 Double activity imaging revealed that haloperidol, clozapine and RGH-1756 share cellular targets in the nucleus accumbens, where 40% of all labeled neurons displayed both c-fos mRNA and c-Fos protein. Clozapine 51-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-176 11166331-5 2001 Double activity imaging revealed that haloperidol, clozapine and RGH-1756 share cellular targets in the nucleus accumbens, where 40% of all labeled neurons displayed both c-fos mRNA and c-Fos protein. Clozapine 51-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 186-191 11248369-2 2001 To confirm the existence of such modulation in c-Fos expression in the trigeminal system, changes in c-Fos expression in the trigeminal nucleus caudalis induced by formalin injection into the rat whisker pad were examined by previously injecting formalin into different areas (contralateral whisker pad, ipsilateral or contralateral forepaw) of the body. Formaldehyde 164-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 11330432-9 2001 In terms of IEP mRNA expression and hepatic regenerative activity, a strong correlation existed between c-fos mRNA expression and [3H]thymidine incorporation (r2 = 0.851, P < 0.01) but not c-jun or c-myc mRNA expression. Tritium 131-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 11330432-13 2001 In conclusion these findings indicate that c-fos mRNA expression 15 mins after PHx correlates with hepatic regenerative activity but not the strength of the regenerative stimulus and that hepatic parenchymal loss of 55-70% must occur prior to the detection of elevated serum bilirubin levels. Bilirubin 275-284 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 11386157-3 2001 Elevation of the c-fos mRNA positive cells number occurred in the hypothalamic" posterior (PHA), lateral (LHA), anterior (AHA), areas dorsomedial (DMH), and ventromedial (VMH) nuclei within 2 hours of the TT administration. Dimenhydrinate 147-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 11386157-4 2001 In 6 hours the c-fos mRNA positive cells number decreased in PHA, LHA, DMH. Dimenhydrinate 71-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 11386157-5 2001 The c-fos mRNA expression was stable in arquate and supraoptic hypothalamic nuclei following either the TT or saline administration. Sodium Chloride 110-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 11248369-3 2001 Formalin injection-evoked c-Fos expression in this nucleus was significantly reduced by previous formalin injection into the contralateral whisker pad or ipsilateral forepaw but not into the contralateral forepaw. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 11248369-3 2001 Formalin injection-evoked c-Fos expression in this nucleus was significantly reduced by previous formalin injection into the contralateral whisker pad or ipsilateral forepaw but not into the contralateral forepaw. Formaldehyde 97-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 11248369-4 2001 The interval between the two injections of formalin that produced a maximal reduction of formalin injection-evoked c-Fos expression was 1 h, and the reduction of c-Fos expression was less when the interval of the two noxious stimuli was longer or shorter than 1 h. These results suggested that noxious stimulus-evoked c-Fos expression in the trigeminal nucleus caudalis is reduced by noxious stimulus applied previously to remote areas, and the reduction is dependent on the area of previous noxious stimulation and interval between the two noxious stimuli. Formaldehyde 43-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 11248369-4 2001 The interval between the two injections of formalin that produced a maximal reduction of formalin injection-evoked c-Fos expression was 1 h, and the reduction of c-Fos expression was less when the interval of the two noxious stimuli was longer or shorter than 1 h. These results suggested that noxious stimulus-evoked c-Fos expression in the trigeminal nucleus caudalis is reduced by noxious stimulus applied previously to remote areas, and the reduction is dependent on the area of previous noxious stimulation and interval between the two noxious stimuli. Formaldehyde 89-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 11172789-0 2001 The elevation of plasma adrenocorticotrophic hormone and expression of c-Fos in hypothalamic paraventricular nucleus by microinjection of neostigmine into the hippocampus in rats: comparison with acute stress responses. Neostigmine 138-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 11172789-3 2001 The patterns of expression of Fos-ir in the PVN after microinjection of neostigmine into the hippocampus were not different from those seen in the two stressful situations. Neostigmine 72-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 11164833-0 2001 Urethane anaesthesia could partly mask antinociceptive effects of non-steroidal anti-inflammatory drugs: a spinal c-Fos protein study. Urethane 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 11223181-7 2001 In electrophoretic mobility shift assays (EMSA), cAMP-inducible nuclear protein complex containing c-Fos formed on the activator protein-1/cAMP responsive element-like site located at -216 to -210 in the promoter of the rat FSH receptor gene. Cyclic AMP 49-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 11223181-7 2001 In electrophoretic mobility shift assays (EMSA), cAMP-inducible nuclear protein complex containing c-Fos formed on the activator protein-1/cAMP responsive element-like site located at -216 to -210 in the promoter of the rat FSH receptor gene. Cyclic AMP 139-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 11172768-5 2001 Immunohistochemical detection of Fos proteins was used as a marker to identify neuronal populations in the thiamin-deficient rat brain affected by glucose loading. Thiamine 107-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 11172768-5 2001 Immunohistochemical detection of Fos proteins was used as a marker to identify neuronal populations in the thiamin-deficient rat brain affected by glucose loading. Glucose 147-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 11172768-6 2001 As thiamin deficiency progressed, the extent and intensity of Fos-like immunoreactivity (FLI) in brain structures typically affected by thiamin deficiency (the thalamus, mammillary bodies, inferior colliculus, vestibular nucleus and inferior olives) were markedly increased when compared to thiamin-replete controls. Thiamine 3-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 11172768-6 2001 As thiamin deficiency progressed, the extent and intensity of Fos-like immunoreactivity (FLI) in brain structures typically affected by thiamin deficiency (the thalamus, mammillary bodies, inferior colliculus, vestibular nucleus and inferior olives) were markedly increased when compared to thiamin-replete controls. Thiamine 136-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 11172768-6 2001 As thiamin deficiency progressed, the extent and intensity of Fos-like immunoreactivity (FLI) in brain structures typically affected by thiamin deficiency (the thalamus, mammillary bodies, inferior colliculus, vestibular nucleus and inferior olives) were markedly increased when compared to thiamin-replete controls. Thiamine 136-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 11172768-7 2001 Glucose loading for 1-3 days further increased the intensity of FLI in these same regions, consistent with a dependence of Fos expression on carbohydrate metabolism as well as on thiamin deficiency. Glucose 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-126 11172768-7 2001 Glucose loading for 1-3 days further increased the intensity of FLI in these same regions, consistent with a dependence of Fos expression on carbohydrate metabolism as well as on thiamin deficiency. Carbohydrates 141-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-126 11172061-7 2001 In both groups, the cocaine S(D), but not the non-reward S(D), elicited strong recovery of responding and increased Fos immunoreactivity in the basolateral amygdala and medial prefrontal cortex (areas Cg1/Cg3). Cocaine 20-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 11172061-8 2001 The response reinstatement and Fos expression induced by the cocaine S(D) were both reversed by selective dopamine D(1) receptor antagonists. Cocaine 61-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 11164833-1 2001 Urethane anaesthesia strongly reduced the peripheral edema (31+/-5 and 96+/-8% reduction of carrageenan-enhanced paw and ankle diameters, respectively; P<0.001 for both) and the spinal c-Fos protein expression (71+/-4% reduction of the number of c-Fos protein-labeled nuclei; P<0.001), 3 h after intraplantar injection of carrageenan in rats. Urethane 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 188-193 11164833-1 2001 Urethane anaesthesia strongly reduced the peripheral edema (31+/-5 and 96+/-8% reduction of carrageenan-enhanced paw and ankle diameters, respectively; P<0.001 for both) and the spinal c-Fos protein expression (71+/-4% reduction of the number of c-Fos protein-labeled nuclei; P<0.001), 3 h after intraplantar injection of carrageenan in rats. Urethane 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 249-254 11164833-1 2001 Urethane anaesthesia strongly reduced the peripheral edema (31+/-5 and 96+/-8% reduction of carrageenan-enhanced paw and ankle diameters, respectively; P<0.001 for both) and the spinal c-Fos protein expression (71+/-4% reduction of the number of c-Fos protein-labeled nuclei; P<0.001), 3 h after intraplantar injection of carrageenan in rats. Carrageenan 92-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 249-254 11164833-3 2001 injection of racemic-flurbiprofen (0.3, 3 and 9 mg/kg) has weaker effects on carrageenan-evoked spinal c-Fos protein expression and peripheral edema than those previously described in the same inflammatory nociceptive model in awake rats. Flurbiprofen 21-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 11164833-3 2001 injection of racemic-flurbiprofen (0.3, 3 and 9 mg/kg) has weaker effects on carrageenan-evoked spinal c-Fos protein expression and peripheral edema than those previously described in the same inflammatory nociceptive model in awake rats. Carrageenan 77-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 11159834-8 2001 In rats infused with the alpha(1)-noradrenergic receptor antagonist, benoxathian, into the supraoptic nucleus before and during iv oxytocin administration, Fos expression in supraoptic neurons was significantly less than that in vehicle controls. benoxathian 69-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-159 11207401-8 2001 Thus, in the spinal dorsal horn of the CCI rats, there was a selective increase in thermal stimulus-induced Fos-LI cells in the superficial dorsal horn after stimulating at near noxious threshold intensities and a non-selective increase in Fos-LI cells in laminae III--IV after both noxious and innocuous thermal stimuli. CCI 39-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 11207401-8 2001 Thus, in the spinal dorsal horn of the CCI rats, there was a selective increase in thermal stimulus-induced Fos-LI cells in the superficial dorsal horn after stimulating at near noxious threshold intensities and a non-selective increase in Fos-LI cells in laminae III--IV after both noxious and innocuous thermal stimuli. CCI 39-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 240-243 11798865-0 2001 [Protective effect of c-fos antisense oligonucleotides on brain damage induced by glutamate]. Oligonucleotides 38-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 11798865-0 2001 [Protective effect of c-fos antisense oligonucleotides on brain damage induced by glutamate]. Glutamic Acid 82-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 11798865-1 2001 OBJECTIVE: To investigate the relation between glutamate neurotoxicity and c-fos gene expression. Glutamic Acid 47-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 11798865-2 2001 METHODS: c-fos antisense oligonucleotides (AS ODN) was injected into the right lateral ventricles of 9 SD rats to block the c-fos gene expression in brain tissue. Oligonucleotides 25-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 11798865-2 2001 METHODS: c-fos antisense oligonucleotides (AS ODN) was injected into the right lateral ventricles of 9 SD rats to block the c-fos gene expression in brain tissue. Oligonucleotides 25-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 11798865-3 2001 c-fos sense oligonucleotides (S ODN) was used a control. Oligonucleotides 12-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 11798865-7 2001 RESULTS: The c-fos AS ODN blocked the c-fos gene expression and reduced the content of both water and sodium in brain tissue and Ca(2+) in symptosome, thus alleviating the morphological damage in neuron. Water 92-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 11798865-7 2001 RESULTS: The c-fos AS ODN blocked the c-fos gene expression and reduced the content of both water and sodium in brain tissue and Ca(2+) in symptosome, thus alleviating the morphological damage in neuron. Sodium 102-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 11798865-10 2001 Blocking the c-fos gene expression could antagonize glutamate neurotoxicity. Glutamic Acid 52-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 11159233-0 2001 Xenon inhibits but N(2)O enhances ketamine-induced c-Fos expression in the rat posterior cingulate and retrosplenial cortices. Nitrous Oxide 19-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 11159233-0 2001 Xenon inhibits but N(2)O enhances ketamine-induced c-Fos expression in the rat posterior cingulate and retrosplenial cortices. Ketamine 34-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 11159233-2 2001 We investigated the effect of xenon, xenon with ketamine, N(2)O, and N(2)O with ketamine on c-Fos expression in the rat posterior cingulate and retrosplenial cortices, a marker of psychotomimetic effects. Ketamine 80-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 11159233-5 2001 The number of Fos-like immunoreactivity positive cells by ketamine IV at a dose of 5 mg/kg under 70% N(2)O (128 +/- 12 cells per 0.5 mm(2)) was significantly more than those under 30% (15 +/- 2 cells per 0.5 mm(2)) and 70% xenon (2 +/- 1 cells per 0.5 mm(2)). Ketamine 58-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 11159233-5 2001 The number of Fos-like immunoreactivity positive cells by ketamine IV at a dose of 5 mg/kg under 70% N(2)O (128 +/- 12 cells per 0.5 mm(2)) was significantly more than those under 30% (15 +/- 2 cells per 0.5 mm(2)) and 70% xenon (2 +/- 1 cells per 0.5 mm(2)). Nitrogen 101-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 11159233-5 2001 The number of Fos-like immunoreactivity positive cells by ketamine IV at a dose of 5 mg/kg under 70% N(2)O (128 +/- 12 cells per 0.5 mm(2)) was significantly more than those under 30% (15 +/- 2 cells per 0.5 mm(2)) and 70% xenon (2 +/- 1 cells per 0.5 mm(2)). Xenon 223-228 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 11159254-7 2001 In addition, continuous methylprednisolone infusion partially reversed nerve injury-evoked Fos expression in the dorsal horns, suggesting that glucocorticoids can inhibit the spinal neuron hyperactivity induced by chronic sciatic nerve transection. Methylprednisolone 24-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-94 11302157-2 2001 Dietary short-chain fructooligosaccharides (Sc-FOS) have been shown to also induce a change in the microflora in the large bowel by promoting an increase in the numbers of Bifidobacterium and Lactobacillus which have beneficial effects on the host. short-chain fructooligosaccharides 8-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 11181452-0 2001 Long-term dehydroepiandrosterone and 16alpha-fluoro-5-androsten-17-one administration enhances DNA synthesis and induces expression of c-fos and c-Ha-ras in a selected population of preneoplastic lesions in liver of diethylnitrosamine-initiated rats. Dehydroepiandrosterone 10-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 11181452-0 2001 Long-term dehydroepiandrosterone and 16alpha-fluoro-5-androsten-17-one administration enhances DNA synthesis and induces expression of c-fos and c-Ha-ras in a selected population of preneoplastic lesions in liver of diethylnitrosamine-initiated rats. 16-fluoro-5-androsten-17-one 37-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 11181452-7 2001 DHEA and FA induced broad increases in lesions with a high LI, which showed a higher number of cells overexpressing c-Ha-ras and/or c-fos than those with a lower LI. Dehydroepiandrosterone 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 11298663-0 2001 Investigations into migraine pathogenesis: time course for effects of m-CPP, BW723C86 or glyceryl trinitrate on appearance of Fos-like immunoreactivity in rat trigeminal nucleus caudalis (TNC). Nitroglycerin 89-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-129 11207816-10 2001 This absence of changes in cocaine-induced Fos-like proteins might result from a compensatory mechanism between the increase in the dopaminergic response and the decrease in the functional activity of dopamine D(1) receptors. Cocaine 27-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 11891731-7 2001 Finally, an increase in c-fos expression was found in irradiated hepatocytes when incubated in the presence of CaCl2. Calcium Chloride 111-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 11272178-4 2001 The general applicability of the approach is exemplified by doxycyclin-(Tet-On) and phorbol 12-myristate 13-acetate-induced (c-fos) promoter activation, with green fluorescent protein (GFP) and red fluorescent protein (DsRed) as semiquantitative and immediate reporters, of transcription activation. Tetradecanoylphorbol Acetate 84-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 11168842-6 2001 Hypertonic saline injection increased the number of cells expressing Fos in the supraoptic nucleus and in the regions anterior and ventral to the third ventricle (AV3V) regions [the organum vasculosum of the lamina terminalis (OVLT) and median preoptic nucleus]. Sodium Chloride 11-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 11120397-0 2001 Opiate withdrawal-induced fos immunoreactivity in the rat extended amygdala parallels the development of conditioned place aversion. Opiate Alkaloids 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 11354804-2 2001 Acute administration of naloxone and chronic administration of morphine changed neither the expression of Fos-LI and NADPH-d positive neurons nor the expression of Fos/NADPH-d double-labeled neurons in the spinal cord of rats. Morphine 63-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 11354804-3 2001 Fos-LI, NADPH-d positive and Fos/NADPH-d double-labeled neurons were increased significantly in number in morphine-withdrawal rats and they were observed in all the laminae of the spinal cord. Morphine 106-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 11354804-3 2001 Fos-LI, NADPH-d positive and Fos/NADPH-d double-labeled neurons were increased significantly in number in morphine-withdrawal rats and they were observed in all the laminae of the spinal cord. Morphine 106-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 11354804-4 2001 Intrathecal injection of L-NA, nNOS antisense oligonucleotides significantly inhibited the expression of Fos-LI in the spinal cord and decreased the scores for morphine-withdrawal symptoms in morphine-withdrawal rats, but not in nNOS-S group. Oligonucleotides 46-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-108 11166695-0 2001 Effects of baclofen on colon inflammation-induced Fos, CGRP and SP expression in spinal cord and brainstem. Baclofen 11-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 11166695-2 2001 The protective effect of baclofen, a selective GABA(B) receptor agonist, on the induced Fos protein increases was determined. Baclofen 25-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 11166695-11 2001 These data suggest that: (1) neurons in regions important for nociceptive transmission, descending inhibitory control and autonomic control are activated by noxious stimulation of the colon, and (2) baclofen specifically reduces Fos expression in the superficial dorsal horn of the spinal cord induced by nociceptive afferent input. Baclofen 199-207 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 229-232 11275410-0 2001 Involvement of nitric oxide in morphine-induced c-Fos expression in the rat striatum. Nitric Oxide 15-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 11275410-0 2001 Involvement of nitric oxide in morphine-induced c-Fos expression in the rat striatum. Morphine 31-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 11275410-1 2001 Induction of expression of immediate-early gene c-Fos in the striatum is a common effect of many drugs of abuse, including morphine. Morphine 123-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 11275410-2 2001 Previous studies have shown that the morphine-mediated c-Fos response is attenuated by antagonists of the N-methyl-D-aspartate (NMDA) subtype of glutamate receptor. Morphine 37-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 11275410-2 2001 Previous studies have shown that the morphine-mediated c-Fos response is attenuated by antagonists of the N-methyl-D-aspartate (NMDA) subtype of glutamate receptor. N-Methylaspartate 106-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 11275410-2 2001 Previous studies have shown that the morphine-mediated c-Fos response is attenuated by antagonists of the N-methyl-D-aspartate (NMDA) subtype of glutamate receptor. N-Methylaspartate 128-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 11275410-5 2001 Because activation of NMDA receptors mediates morphine-induced c-Fos expression, we tested the hypothesis that activation of nNOS is involved in this cascade. Morphine 46-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 11275410-8 2001 Morphine induced c-Fos expression in the striatum, cerebral cortex, and midline/intralaminar nuclei of thalamus. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 11150488-4 2001 No differences were observed between these administration paradigms; both single and subchronic PCP exposure enhanced amphetamine-induced c-Fos in the striatum, decreased c-Fos in the prefrontal cortex, and decreased the number of cage-crossings. Phencyclidine 96-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 11150488-4 2001 No differences were observed between these administration paradigms; both single and subchronic PCP exposure enhanced amphetamine-induced c-Fos in the striatum, decreased c-Fos in the prefrontal cortex, and decreased the number of cage-crossings. Phencyclidine 96-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-176 11150488-4 2001 No differences were observed between these administration paradigms; both single and subchronic PCP exposure enhanced amphetamine-induced c-Fos in the striatum, decreased c-Fos in the prefrontal cortex, and decreased the number of cage-crossings. Amphetamine 118-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 11150488-5 2001 However, the observation that PCP pretreatment affected c-Fos induction in the same manner observed previously while having an opposite effect on amphetamine-induced behavior suggests that these behavioral and neurochemical effects are dissociated. Phencyclidine 30-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 11160452-0 2001 Environmental novelty differentially affects c-fos mRNA expression induced by amphetamine or cocaine in subregions of the bed nucleus of the stria terminalis and amygdala. Amphetamine 78-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 11160452-0 2001 Environmental novelty differentially affects c-fos mRNA expression induced by amphetamine or cocaine in subregions of the bed nucleus of the stria terminalis and amygdala. Cocaine 93-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 11160452-2 2001 Here we report that environmental context differentially affects patterns of amphetamine- and cocaine-induced c-fos mRNA expression in the bed nucleus of the stria terminalis (BST) and amygdala of male rats. Amphetamine 77-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 11160452-2 2001 Here we report that environmental context differentially affects patterns of amphetamine- and cocaine-induced c-fos mRNA expression in the bed nucleus of the stria terminalis (BST) and amygdala of male rats. Cocaine 94-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 11160452-5 2001 In the basolateral and lateral amygdala, amphetamine or cocaine at home or exposure to novelty induced c-fos mRNA. Amphetamine 41-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 11160452-5 2001 In the basolateral and lateral amygdala, amphetamine or cocaine at home or exposure to novelty induced c-fos mRNA. Cocaine 56-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 11160452-6 2001 When amphetamine or cocaine was given in a novel environment the c-fos mRNA response was significantly enhanced. Amphetamine 5-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 11160452-6 2001 When amphetamine or cocaine was given in a novel environment the c-fos mRNA response was significantly enhanced. Cocaine 20-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 11160452-7 2001 In the central nucleus of the amygdala (CEA) and oval subnucleus of the BST (BSTov), amphetamine administration at home produced a robust increase in c-fos mRNA expression, whereas exposure to novelty had little effect. Amphetamine 85-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-155 11160452-8 2001 In contrast to other brain regions examined, the c-fos mRNA response to amphetamine in a novel versus home environment was significantly smaller. Amphetamine 72-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 11160452-9 2001 In both "home" and "novel" amphetamine groups, c-fos mRNA in the BSTov and CEA was predominantly expressed in enkephalin-containing cells; coexpression with corticotropin-releasing hormone was rare. Amphetamine 27-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 11146065-0 2001 How central is nitric oxide (NO) to the activation of c-fos in spinal neurones following noxious peripheral stimulation in the rat? Nitric Oxide 15-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 11123204-6 2001 Actinomycin D also prevented the perfusion-induced increase in c-fos and c-jun mRNA abundance but did not affect glucose uptake rate and SGLT-1 mRNA abundance. Dactinomycin 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 11124163-3 2001 Systemic exposure to LPS elicited a significant activation of c-Fos in neurons in the nucleus of the solitary tract (NST) and area postrema of all thiobutabarbital-anesthetized rats examined, regardless of the integrity of their vagal nerves. thiobutabarbital 147-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 11173221-10 2001 Furthermore, cFos activation by 8-OH-DPAT was blocked by pimozide--a 5-HT(7) antagonist. 8-Hydroxy-2-(di-n-propylamino)tetralin 32-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-17 11173221-10 2001 Furthermore, cFos activation by 8-OH-DPAT was blocked by pimozide--a 5-HT(7) antagonist. Pimozide 57-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-17 11169614-7 2001 Present results suggest that Fos expression in various brainstem areas was induced by reduced oxygen tension in the ambient air at high altitude. Oxygen 94-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 11274784-3 2001 After administration of 30mg/kg or 100mg/kg of L-DOPA, rats subjected to unilateral dopaminergic denervation showed intense contraversive rotation and a high density of Fos-immunoreactive nuclei throughout the denervated striatum, with no significant induction of Fos in the intact striatum. Levodopa 47-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-172 11274784-3 2001 After administration of 30mg/kg or 100mg/kg of L-DOPA, rats subjected to unilateral dopaminergic denervation showed intense contraversive rotation and a high density of Fos-immunoreactive nuclei throughout the denervated striatum, with no significant induction of Fos in the intact striatum. Levodopa 47-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 264-267 11274785-2 2001 The full D-1 receptor agonist, SKF-82958 (0.05, 0.1, 0.5 and 1 mg/kg, s.c., 30 min), dose-dependently induced c-fos messenger RNA in naive rat striatum as determined by northern blot analysis. 1,2,3,4-tetrahydroisoquinoline-7-sulfonamide 31-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 11274784-4 2001 Injection of the central aromatic L-amino-acid decarboxylase inhibitor NSD-1015 30min before and 15min after the injection of L-DOPA suppressed the rotational behavior and the striatal induction of Fos. 3-hydroxybenzylhydrazine 71-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 198-201 11274784-4 2001 Injection of the central aromatic L-amino-acid decarboxylase inhibitor NSD-1015 30min before and 15min after the injection of L-DOPA suppressed the rotational behavior and the striatal induction of Fos. Levodopa 126-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 198-201 11274784-6 2001 Serotonergic denervation led to slight and statistically non-significant decrease in the rotational behavior and Fos expression induced by high doses of L-DOPA (100mg/kg) in the dopamine-denervated striatum, but totally suppressed the rotational behavior and Fos expression induced by low doses of L-DOPA (30mg/kg). Levodopa 153-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-116 11301212-1 2001 We examined the effects of systemic administration of a GABA(B) receptor agonist, baclofen, or antagonist, phaclofen, on the expression of c-Fos protein induced 3h after electrical stimulation of the trigeminal ganglion at low (0.1 mA) or high intensities (1.0 mA) in the urethane-anesthetized rat. gamma-Aminobutyric Acid 56-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 11301212-1 2001 We examined the effects of systemic administration of a GABA(B) receptor agonist, baclofen, or antagonist, phaclofen, on the expression of c-Fos protein induced 3h after electrical stimulation of the trigeminal ganglion at low (0.1 mA) or high intensities (1.0 mA) in the urethane-anesthetized rat. Baclofen 82-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 11301212-1 2001 We examined the effects of systemic administration of a GABA(B) receptor agonist, baclofen, or antagonist, phaclofen, on the expression of c-Fos protein induced 3h after electrical stimulation of the trigeminal ganglion at low (0.1 mA) or high intensities (1.0 mA) in the urethane-anesthetized rat. phaclofen 107-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 11301212-1 2001 We examined the effects of systemic administration of a GABA(B) receptor agonist, baclofen, or antagonist, phaclofen, on the expression of c-Fos protein induced 3h after electrical stimulation of the trigeminal ganglion at low (0.1 mA) or high intensities (1.0 mA) in the urethane-anesthetized rat. Tritium 161-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 11301212-1 2001 We examined the effects of systemic administration of a GABA(B) receptor agonist, baclofen, or antagonist, phaclofen, on the expression of c-Fos protein induced 3h after electrical stimulation of the trigeminal ganglion at low (0.1 mA) or high intensities (1.0 mA) in the urethane-anesthetized rat. Urethane 272-280 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 11274784-6 2001 Serotonergic denervation led to slight and statistically non-significant decrease in the rotational behavior and Fos expression induced by high doses of L-DOPA (100mg/kg) in the dopamine-denervated striatum, but totally suppressed the rotational behavior and Fos expression induced by low doses of L-DOPA (30mg/kg). Dopamine 178-186 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-116 11301212-2 2001 In saline-treated rats, 10 min stimulation of the trigeminal ganglion induced c-Fos-immunopositive neurons throughout the full extent of the ipsilateral superficial layers of the trigeminal nucleus caudalis, and dorsal or dorsomedial part of the nuclei rostral to obex (trigeminal nucleus principalis, dorsomedial nucleus of trigeminal nucleus oralis and dorsomedial nucleus of trigeminal nucleus interpolaris). Sodium Chloride 3-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 11311795-4 2001 Both the re-expression of angiotensin II subtype 1 receptor messenger RNA and restoration of pressor response to angiotensin II after baroreceptor activation were significantly blunted by bilateral application into the nucleus tractus solitarii of an antisense oligonucleotide (50 pmol) that targets against the initiation codon of c-fos messenger RNA. Oligonucleotides 261-276 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 332-337 11301212-5 2001 baclofen and stimulated at 0.1 mA, the numbers of Fos-positive neurons in all the trigeminal sensory nuclei were significantly decreased compared to saline-treated controls. Baclofen 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 11301212-6 2001 After stimulation at 1.0 mA in rats treated with baclofen, the numbers of Fos-positive neurons in all the trigeminal sensory nuclei were also significantly decreased. Baclofen 49-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 11301212-8 2001 phaclofen and stimulated at 1.0 mA, the numbers of Fos-positive neurons were significantly increased in all the trigeminal sensory nuclei. phaclofen 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 11301212-9 2001 However, after stimulation at 0.1 mA in rats treated with phaclofen, the numbers of Fos-positive neurons were significantly decreased in the superficial layers and magnocellular zone of trigeminal nucleus caudalis and dorsomedial nucleus of trigeminal nucleus oralis. phaclofen 58-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 11440804-1 2001 The immediate-early gene product Fos is differentially induced in the rat brain by the antipsychotic drugs haloperidol and clozapine. Haloperidol 107-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 11440804-1 2001 The immediate-early gene product Fos is differentially induced in the rat brain by the antipsychotic drugs haloperidol and clozapine. Clozapine 123-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 11440804-2 2001 It is often claimed that although both drugs induce Fos in the nucleus accumbens, haloperidol but not clozapine increases Fos-like immunoreactivity in the striatum, whereas clozapine but not haloperidol increases Fos-like immunoreactivity in prefrontal cortex. Haloperidol 82-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 11440804-2 2001 It is often claimed that although both drugs induce Fos in the nucleus accumbens, haloperidol but not clozapine increases Fos-like immunoreactivity in the striatum, whereas clozapine but not haloperidol increases Fos-like immunoreactivity in prefrontal cortex. Haloperidol 82-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 11440804-4 2001 In the present study, we compared the effects of low doses of haloperidol (0.1 mg/kg) and clozapine (5 mg/kg) on Fos-like immunoreactivity in rats which were either behaviorally naive, exposed to a novel environment or tested for two-way active avoidance. Haloperidol 62-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-116 11440804-4 2001 In the present study, we compared the effects of low doses of haloperidol (0.1 mg/kg) and clozapine (5 mg/kg) on Fos-like immunoreactivity in rats which were either behaviorally naive, exposed to a novel environment or tested for two-way active avoidance. Clozapine 90-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-116 11440804-5 2001 We determined that haloperidol increased Fos in the striatum and nucleus accumbens regardless of testing condition whereas clozapine markedly reduced the induction of Fos by behavioral testing in these regions; moreover, haloperidol dramatically increased prefrontal cortical Fos expression in animals placed in a novel environment, but not in testing-naive controls. Haloperidol 19-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 11440804-5 2001 We determined that haloperidol increased Fos in the striatum and nucleus accumbens regardless of testing condition whereas clozapine markedly reduced the induction of Fos by behavioral testing in these regions; moreover, haloperidol dramatically increased prefrontal cortical Fos expression in animals placed in a novel environment, but not in testing-naive controls. Clozapine 123-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 167-170 11440804-5 2001 We determined that haloperidol increased Fos in the striatum and nucleus accumbens regardless of testing condition whereas clozapine markedly reduced the induction of Fos by behavioral testing in these regions; moreover, haloperidol dramatically increased prefrontal cortical Fos expression in animals placed in a novel environment, but not in testing-naive controls. Clozapine 123-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 167-170 11490180-1 2001 In the search for differential mechanisms underlying clozapine"s superior antipsychotic efficacy, the purinergic system has been considered, since an antagonist of the adenosine receptor A(2A) was shown to block clozapine acute effects on c-fos expression in rat striatum. Clozapine 53-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 239-244 11530233-2 2001 Since it has been observed that heme oxygenase inhibitors reduce formalin-induced pain behaviors in mice and rats, we attempted to determine if this analgesic effect was reflected in a reduction in formalin-induced spinal cord Fos expression, an index of neuronal activation. Formaldehyde 198-206 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 227-230 11564427-0 2001 NMDA or non-NMDA receptor antagonists attenuate increased Fos expression in spinal dorsal horn GABAergic neurons after intradermal injection of capsaicin in rats. N-Methylaspartate 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 11564427-0 2001 NMDA or non-NMDA receptor antagonists attenuate increased Fos expression in spinal dorsal horn GABAergic neurons after intradermal injection of capsaicin in rats. Capsaicin 144-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 11564427-5 2001 Western blots showed that Fos protein was increased on the ipsilateral side in spinal cord tissue 0.5 h after capsaicin injection. Capsaicin 110-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 11564427-6 2001 Pretreatment with AP7 or CNQX caused a decrease in capsaicin-induced Fos expression. ap7 18-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 11564427-6 2001 Pretreatment with AP7 or CNQX caused a decrease in capsaicin-induced Fos expression. 6-Cyano-7-nitroquinoxaline-2,3-dione 25-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 11564427-6 2001 Pretreatment with AP7 or CNQX caused a decrease in capsaicin-induced Fos expression. Capsaicin 51-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 11564427-7 2001 Immunofluorescence double labeling showed that the proportion of Fos-positive GABAergic neuronal profiles was significantly increased following capsaicin injection (48.8+/-4.8%) compared to the vehicle injection (23.8+/-5.1%) in superficial laminae on the ipsilateral side in lumbosacral spinal cord (P<0.05). Capsaicin 144-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 11564427-8 2001 However, when the spinal cord was pretreated with AP7 (5 microg) or CNQX (0.2 microg), only 9.1+/-0.6% or 7.1+/-0.8% of GABA-immunoreactive neuronal profiles were stained for Fos following capsaicin injection. ap7 50-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 175-178 11564427-8 2001 However, when the spinal cord was pretreated with AP7 (5 microg) or CNQX (0.2 microg), only 9.1+/-0.6% or 7.1+/-0.8% of GABA-immunoreactive neuronal profiles were stained for Fos following capsaicin injection. 6-Cyano-7-nitroquinoxaline-2,3-dione 68-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 175-178 11564427-9 2001 The blockade of the capsaicin-evoked Fos staining was dose-dependent. Capsaicin 20-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 11591463-6 2001 However, the self-stimulation-induced expression of Fos was restricted mostly to GABA-, but not choline acetyltransferase-, immunostained cells. gamma-Aminobutyric Acid 81-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 11311795-5 2001 Control pretreatment with the corresponding sense oligonucleotide (50 pmol), or an antisense c-fos oligonucleotide that targets against a different portion of the coding sequence of the c-fos messenger RNA (50 pmol), was ineffective. Oligonucleotides 99-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 11682155-0 2001 Expression of c-Fos, Fos B, Jun B, and Zif268 transcription factor proteins in rat barrel cortex following apomorphine-evoked whisking behavior. Apomorphine 107-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 11672604-4 2001 To determine whether the ability of the D2 receptor antagonist, sulpiride, to induce Fos in rat pallidal neurons is mediated by D2-like receptors in striatum or globus pallidus, intrastriatal or intrapallidal sulpiride infusions were conducted. Sulpiride 64-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 11672604-7 2001 Intrastriatal infusions of (-)-sulpiride (10-200 ng) dose-dependently increased the number of striatal cells expressing Fos; and the Fos-immunoreactive striatal cells were D2 receptor mRNA-expressing, the same population in which systemic D2 receptor antagonists induce Fos. Sulpiride 27-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-123 11672604-7 2001 Intrastriatal infusions of (-)-sulpiride (10-200 ng) dose-dependently increased the number of striatal cells expressing Fos; and the Fos-immunoreactive striatal cells were D2 receptor mRNA-expressing, the same population in which systemic D2 receptor antagonists induce Fos. Sulpiride 27-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-136 11672604-7 2001 Intrastriatal infusions of (-)-sulpiride (10-200 ng) dose-dependently increased the number of striatal cells expressing Fos; and the Fos-immunoreactive striatal cells were D2 receptor mRNA-expressing, the same population in which systemic D2 receptor antagonists induce Fos. Sulpiride 27-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-136 11672604-8 2001 Intrastriatal infusions of high (5 microg), but not low (10-200 ng), (-)-sulpiride doses also induced Fos in globus pallidus cells but the sulpiride appeared to spread to the globus pallidus. Sulpiride 69-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 11672604-8 2001 Intrastriatal infusions of high (5 microg), but not low (10-200 ng), (-)-sulpiride doses also induced Fos in globus pallidus cells but the sulpiride appeared to spread to the globus pallidus. Sulpiride 73-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 11672604-9 2001 Direct intrapallidal infusions of (-)-sulpiride (50-100 ng) dose-dependently induced Fos in globus pallidus with minimal influence on striatum or other basal ganglia structures. Sulpiride 34-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 11719001-25 2001 Rats receiving low dose corticosterone showed increased Fos-b expression following 9 days stress in the lateral septum and in the dorsal and medial parts of the paraventricular nucleus compared to either control, stressed rats or those receiving the higher corticosterone dose and repeated stress. Corticosterone 24-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 11720788-8 2001 CGS21680 also induced Fos in the shell and core of the nucleus accumbens and in the lateral subdivision of the central nucleus of the amygdala. 2-(4-(2-carboxyethyl)phenethylamino)-5'-N-ethylcarboxamidoadenosine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-25 11720788-10 2001 In contrast to CGS21680, infusion of N(6)-cyclopentyladenosine into the subarachnoid space produced only a small decrease in rapid eye movement sleep, and the pattern of Fos expression induced by N(6)-cyclopentyladenosine was notable only for decreased Fos in regions near the infusion site. N(6)-cyclopentyladenosine 37-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 253-256 11720788-10 2001 In contrast to CGS21680, infusion of N(6)-cyclopentyladenosine into the subarachnoid space produced only a small decrease in rapid eye movement sleep, and the pattern of Fos expression induced by N(6)-cyclopentyladenosine was notable only for decreased Fos in regions near the infusion site. N(6)-cyclopentyladenosine 196-221 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-173 11720788-10 2001 In contrast to CGS21680, infusion of N(6)-cyclopentyladenosine into the subarachnoid space produced only a small decrease in rapid eye movement sleep, and the pattern of Fos expression induced by N(6)-cyclopentyladenosine was notable only for decreased Fos in regions near the infusion site. N(6)-cyclopentyladenosine 196-221 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 253-256 11738500-0 2001 Brainstem and hypothalamic areas activated by tissue hypoxia: Fos-like immunoreactivity induced by carbon monoxide inhalation in the rat. Carbon Monoxide 99-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 11738500-3 2001 We addressed this issue, mapping Fos expression in adult conscious rats subjected to tissue hypoxia elicited by carbon monoxide inhalation, under conditions of minimal activation of arterial chemoreceptors. Carbon Monoxide 112-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 11744251-0 2001 Basal increase in c-Fos-like expression in superior colliculus of Royal College of Surgeons dystrophic rats can be abolished by intraocular injection of tetrodotoxin. Tetrodotoxin 153-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 11166125-0 2001 Glutamatergic regulation of haloperidol-induced c-fos expression in the rat striatum and nucleus accumbens. Haloperidol 28-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 11166125-1 2001 Acute administration of haloperidol induces the expression of the immediate-early gene c-fos in the striatum and nucleus accumbens via dopamine D(2) receptor antagonism. Haloperidol 24-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 11166125-3 2001 Indeed, haloperidol-induced c-fos expression is dependent on NMDA receptor activation in the dorsolateral part of the striatum. Haloperidol 8-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 11166125-4 2001 However, the role that NMDA receptors play in haloperidol-induced c-fos expression in other functionally distinct areas of the striatum and nucleus accumbens has not yet been established. Haloperidol 46-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 11166125-5 2001 Therefore, in the present study the entire rostrocaudal extent of the rat striatum and nucleus accumbens was examined to determine the role that NMDA receptors play in haloperidol-induced c-fos expression. Haloperidol 168-179 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 188-193 11166125-6 2001 Pretreatment with MK-801, a non-competitive antagonist of NMDA receptors, significantly reduced the number of neurons showing c-fos immunoreactivity in the rostral aspect of the dorsolateral striatum and the entire rostrocaudal extent of the ventrolateral striatum following an acute injection of haloperidol. Dizocilpine Maleate 18-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 11166125-6 2001 Pretreatment with MK-801, a non-competitive antagonist of NMDA receptors, significantly reduced the number of neurons showing c-fos immunoreactivity in the rostral aspect of the dorsolateral striatum and the entire rostrocaudal extent of the ventrolateral striatum following an acute injection of haloperidol. Haloperidol 297-308 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 11166125-8 2001 Similarly, MK-801 pretreatment significantly suppressed the number of neurons expressing c-fos immunoreactivity following haloperidol injection in the entire rostrocaudal extent of the shell region of nucleus accumbens, but not in the core region. Dizocilpine Maleate 11-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 11166125-8 2001 Similarly, MK-801 pretreatment significantly suppressed the number of neurons expressing c-fos immunoreactivity following haloperidol injection in the entire rostrocaudal extent of the shell region of nucleus accumbens, but not in the core region. Haloperidol 122-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 11166125-9 2001 The results indicate that haloperidol-induced c-fos expression is dependent on NMDA receptors only in the rostral aspect of the dorsolateral striatum and the rostrocaudal extent of the ventrolateral striatum, the areas involved in motor function. Haloperidol 26-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 11166125-11 2001 Furthermore, the attenuation of the haloperidol-induced c-fos expression by MK-801 was restricted to the nucleus accumbens shell, an area often implicated in the therapeutic effect of haloperidol. Haloperidol 36-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 11166125-11 2001 Furthermore, the attenuation of the haloperidol-induced c-fos expression by MK-801 was restricted to the nucleus accumbens shell, an area often implicated in the therapeutic effect of haloperidol. Dizocilpine Maleate 76-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 11166125-11 2001 Furthermore, the attenuation of the haloperidol-induced c-fos expression by MK-801 was restricted to the nucleus accumbens shell, an area often implicated in the therapeutic effect of haloperidol. Haloperidol 184-195 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 11166470-4 2001 IG HCl caused many neurons in the NTS and some neurons in the AP to express c-fos mRNA. Hydrochloric Acid 3-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 11166470-7 2001 In contrast, the c-fos mRNA response in the AP was significantly augmented by MK-801, an action that was prevented by coadministration of GR-205,171 plus SR-144,190. Dizocilpine Maleate 78-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 11179601-1 2001 We studied whether TRH receptor 1 (TRH-R1) antisense oligodeoxynucleotides injected intracisternally, impaired acute cold-induced c-Fos expression in the brain and vagally mediated stimulation of gastric emptying in conscious rats. Oligodeoxyribonucleotides 53-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 11860963-0 2001 [The effect of alcohol on c-fos gene expression in rat embryo neuroglial]. Alcohols 15-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 11860963-4 2001 RESULTS: Changes in c-fos gene expression induced by alcohol and acetaldehyde was time and dose dependent. Alcohols 53-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 11860963-4 2001 RESULTS: Changes in c-fos gene expression induced by alcohol and acetaldehyde was time and dose dependent. Acetaldehyde 65-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 11860963-5 2001 After 1 hr exposure, alcohol and acetaldehyde affected c-fos gene expression in two kinds of neuralglia. Alcohols 21-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 11860963-5 2001 After 1 hr exposure, alcohol and acetaldehyde affected c-fos gene expression in two kinds of neuralglia. Acetaldehyde 33-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 11860963-7 2001 CONCLUSIONS: Alcohol and acetaldehyde cause abnormal increase of c-fos gene expression in astrocytes and oligodendrocytes. Alcohols 13-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 11860963-7 2001 CONCLUSIONS: Alcohol and acetaldehyde cause abnormal increase of c-fos gene expression in astrocytes and oligodendrocytes. Acetaldehyde 25-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 12516397-0 2000 [Potential anxiolytic role of c-fos antisense oligonucleotide in social-defeated rats]. Oligonucleotides 46-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 12516397-1 2000 OBJECTIVES: This study was to explore the therapeutic potential of c-fos antisense phosphorothioate oligonucleotide and the possible molecular mechanism. Phosphorothioate Oligonucleotides 83-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 12516397-2 2000 METHODS: On the basis of the sequence of c-fos mRNA, two 15-mer phosphorothioated oligodeoxynucleotide(ODN) were synthesized. phosphorothioated oligodeoxynucleotide 64-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 12516397-2 2000 METHODS: On the basis of the sequence of c-fos mRNA, two 15-mer phosphorothioated oligodeoxynucleotide(ODN) were synthesized. odn 103-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 12516397-11 2000 Antisense oligodeoxynucleotide significantly decreased Fos positive nucleus in paraventricularis hypothalami nucleus. Oligodeoxyribonucleotides 10-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 11125002-3 2000 In rats, Fos immunoreactivity was examined in brainstem noradrenergic nuclei after exposure to nitrous oxide. Nitrous Oxide 95-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 11090977-0 2000 Fos activation and upregulation of nicotinamide adenine dinucleotide phosphate diaphorase in the rat pituitary by acute capsaicin injection. Capsaicin 120-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 11090977-2 2000 Compared with vehicle, capsaicin significantly activated Fos expression in the anterior and intermediate lobes. Capsaicin 23-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 11090977-4 2000 Pretreatment of the animals with a specific NO synthase inhibitor, N(omega)-nitro-L-arginine methyl ester (L-NAME), significantly attenuated the capsaicin-induced Fos expression in the anterior and intermediate lobes. NG-Nitroarginine Methyl Ester 67-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 11090977-4 2000 Pretreatment of the animals with a specific NO synthase inhibitor, N(omega)-nitro-L-arginine methyl ester (L-NAME), significantly attenuated the capsaicin-induced Fos expression in the anterior and intermediate lobes. NG-Nitroarginine Methyl Ester 107-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 11090977-4 2000 Pretreatment of the animals with a specific NO synthase inhibitor, N(omega)-nitro-L-arginine methyl ester (L-NAME), significantly attenuated the capsaicin-induced Fos expression in the anterior and intermediate lobes. Capsaicin 145-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 11682155-1 2001 Apomorphine-evoked expression of transcription factor proteins: c-Fos, Fos B, Jun B, and Zif268 (also named Krox-24, NGFI-A, Egr-1), was investigated in rat somatosensory (barrel) cortex. Apomorphine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 11682155-6 2001 In contrast, apomorphine (5.0 mg/kg) evoked marked c-Fos elevation, less pronounced changes in Jun B and Zif268 and no change in Fos B. Apomorphine 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 11682155-6 2001 In contrast, apomorphine (5.0 mg/kg) evoked marked c-Fos elevation, less pronounced changes in Jun B and Zif268 and no change in Fos B. Apomorphine 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 11682155-7 2001 The greatest apomorphine-evoked c-Fos accumulation was observed in layers IV and V/VI of non-deprived barrel cortex and was not significantly influenced by MK-801 injection at 0.1 mg/kg. Apomorphine 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 11682155-8 2001 A higher dose of MK-801 (1.0 mg/kg) produced abnormalities in locomotor behavior and diminished c-Fos levels on the non-deprived side to the ones observed in the sensory stimulus-deprived cortex. Dizocilpine Maleate 17-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 11682167-5 2001 DNQX, but not AP-5, significantly reduced the total number of Fos-like immunoreactive neurons evoked by muscle inflammation. FG 9041 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 11682167-6 2001 In contrast, superficial tissue inflammation evoked expression of Fos-like immunoreactive neurons in the medial portions of laminae I-II(outer) and V-VI of the ipsilateral dorsal horn at the spinal L(4)-L(5) that was blocked by AP-5, but not by DNQX. 2-amino-5-phosphopentanoic acid 228-232 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 11682167-6 2001 In contrast, superficial tissue inflammation evoked expression of Fos-like immunoreactive neurons in the medial portions of laminae I-II(outer) and V-VI of the ipsilateral dorsal horn at the spinal L(4)-L(5) that was blocked by AP-5, but not by DNQX. FG 9041 245-249 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 11085902-9 2000 Gp120 induced the expression of Fos protein in both the parvo- and the magnocellular PVN, which was significantly attenuated by l-NMMA 10(-6) nM/L (P < 0.001 vs gp120 alone). omega-N-Methylarginine 128-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 11074189-8 2000 Rats with seizures induced by PAG carbachol microinjections exhibited dense c-fos-like immunoreactivity in the dentate gyrus but not the CA(1) or CA(3) regions, amygdala, piriform cortex, perirhinal cortex or hypothalamus. Carbachol 34-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 11115071-12 2000 Moreover, geldanamycin interfered with mitogen-induced phosphorylation of extracellular signal-regulated kinase and transcription of c-fos and Egr-1, but not with transactivation of STAT1 transcription factor. geldanamycin 10-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 11099700-6 2000 L-NAME potentiated PCP-induced behaviours and c-fos expression in many brain regions. NG-Nitroarginine Methyl Ester 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 11122358-0 2000 Mapping of c-fos gene expression in the brain during morphine dependence and precipitated withdrawal, and phenotypic identification of the striatal neurons involved. Morphine 53-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 11122358-2 2000 Using in situ hybridization, we examined the effects of chronic morphine treatment and withdrawal on c-fos mRNA in the rat brain, and particularly within identified striatal neurons. Morphine 64-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 11122358-4 2000 Placebo animals and morphine-dependent rats showed a very weak c-fos mRNA expression in all the structures studied. Morphine 20-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 11122358-5 2000 Our study emphasized the spatial variations in c-fos mRNA expression, and also revealed a peak expression of c-fos mRNA at 1 h after naltrexone-precipitated withdrawal in the projection areas of dopaminergic neurons, noradrenergic neurons and in several regions expressing opiate receptors. Naltrexone 133-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 11122358-6 2000 Interestingly, morphine withdrawal induces c-fos mRNA expression in the two efferent populations of the striatum (i.e. striatonigral and striatopallidal neurons) both in the caudate putamen and nucleus accumbens. Morphine 15-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 11122358-9 2000 On the contrary, c-fos induction in striatonigral neurons during withdrawal seems to involve a more complex regulation like opioid-dopamine interactions via the mu opioid receptor and the D1 dopamine receptor coexpressed on this neuronal population or the implication of other neurotransmitter systems. Dopamine 131-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 11102493-1 2000 Administration of the hallucinogenic 5-HT(2A/2C) agonist 1-[2, 5-dimethoxy-4-iodophenyl]-2-aminopropane (DOI) induces expression of Fos protein in the cerebral cortex. 4-iodo-2,5-dimethoxyphenylisopropylamine 57-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-135 11102493-9 2000 These data suggest that DOI activates 5-HT(2A) receptors on thalamocortical neurons and thereby increases glutamate release, which in turn drives Fos expression in cortical neurons through an AMPA receptor-dependent mechanism. Glutamic Acid 106-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-149 11164559-4 2000 Numerous numbers of Fos-immunoreactive (Fos-ir) cells were found in the spVc and PAG by stimulation of the tooth pulp with acetic acid or saline. Acetic Acid 123-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 11164559-4 2000 Numerous numbers of Fos-immunoreactive (Fos-ir) cells were found in the spVc and PAG by stimulation of the tooth pulp with acetic acid or saline. Acetic Acid 123-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 11164559-4 2000 Numerous numbers of Fos-immunoreactive (Fos-ir) cells were found in the spVc and PAG by stimulation of the tooth pulp with acetic acid or saline. Sodium Chloride 138-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 11164559-4 2000 Numerous numbers of Fos-immunoreactive (Fos-ir) cells were found in the spVc and PAG by stimulation of the tooth pulp with acetic acid or saline. Sodium Chloride 138-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 11211055-0 2000 Convulsive effects of thiophene, a heterocyclic hydrocarbon: behavioral, electrographic and c-Fos immunocytochemical studies. Thiophenes 22-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 11239672-1 2000 Expression of c-fos is increased in the central amygdaloid nucleus (CE) of rats ingesting a diet with a severely imbalanced essential amino acid profile (IMB), at a time associated with development of a conditioned taste aversion (CTA). Amino Acids, Essential 124-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 11069971-5 2000 We found that 75 mg/kg modafinil increased Fos immunoreactivity in the tuberomammillary nucleus (TMN) and in orexin (hypocretin) neurons of the perifornical area, two cell groups implicated in the regulation of wakefulness. Modafinil 23-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 11146420-6 2000 Within the c-fos promoter the serum response element and the cAMP response element play a permissive role in Ca(2+)- and cAMP-enhanced transcription of intron containing reporter genes. Cyclic AMP 61-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 11146420-6 2000 Within the c-fos promoter the serum response element and the cAMP response element play a permissive role in Ca(2+)- and cAMP-enhanced transcription of intron containing reporter genes. Cyclic AMP 121-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 11069971-6 2000 This low dose of modafinil also increased the number of Fos-immunoreactive (Fos-IR) neurons in the lateral subdivision of the central nucleus of the amygdala. Modafinil 17-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 11069971-6 2000 This low dose of modafinil also increased the number of Fos-immunoreactive (Fos-IR) neurons in the lateral subdivision of the central nucleus of the amygdala. Modafinil 17-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 11070178-0 2000 Limited participation of 5-HT(1A) and 5-HT(2A/2C) receptors in the clozapine-induced Fos-protein expression in rat forebrain regions. Clozapine 67-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 11070178-1 2000 Through the development of tolerance following long-term clozapine treatment, we investigated whether 5-HT(1A) and 5-HT(2A/2C) receptors participate in the clozapine-induced Fos-protein expression in the rat forebrain. Clozapine 156-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 174-177 11070178-7 2000 In the hypothalamic paraventricular nucleus, both clozapine and DOI/8-OH-DPAT induced a remarkably high number of Fos-positive nuclei. Clozapine 50-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 11054694-1 2000 A c-Fos study in the rat central nervous system after intravenous injection of naloxone or naloxone-methiodide. Naloxone 79-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 2-7 11070178-7 2000 In the hypothalamic paraventricular nucleus, both clozapine and DOI/8-OH-DPAT induced a remarkably high number of Fos-positive nuclei. 8-Hydroxy-2-(di-n-propylamino)tetralin 68-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 11054694-1 2000 A c-Fos study in the rat central nervous system after intravenous injection of naloxone or naloxone-methiodide. N-methylnaloxone 91-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 2-7 11070178-8 2000 Long-term clozapine pretreatment attenuated the acutely induced Fos expression of the 5-HT receptor agonists in the nucleus accumbens core, the dorsomedial and ventromedial parts of the striatum and the lateral septum, indicating (partial) common sites of action of the agents in these brain regions. Clozapine 10-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 11070178-9 2000 No tolerance was found in the nucleus accumbens shell and the hypothalamic paraventricular nucleus and the central amygdala, suggesting that the clozapine-induced Fos responses, though distinct in these regions, are independent of 5-HT receptors. Clozapine 145-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 11070178-12 2000 The present results indicate that the clozapine-induced patterns of Fos expression in the rat forebrain can only be in part attributed to an interaction with 5-HT(1A/2A/2C) receptors. Clozapine 38-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 11072104-5 2000 The alpha(2)-AR agonists, clonidine and UK14304 also had no effect on basal IEG levels, while blockade of alpha(2)-ARs by methoxyidazoxan significantly increased NGFI-A and c-fos expression, but decreased c-jun mRNA levels. methoxyidazoxan 122-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 173-178 11072104-4 2000 Activation of alpha(1)-ARs by NVI-085, or beta-ARs by salbutamol, increased cortical NGFI-A, c-jun and c-fos mRNA levels, whereas inhibition of alpha(1)-ARs by prazosin, or beta-ARs by propranolol, had no marked effect. nvi 30-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 11072104-8 2000 Prazosin reduced FCL(SD)-induced elevations of c-jun and c-fos, but not NGFI-A, mRNA. Prazosin 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 11072104-8 2000 Prazosin reduced FCL(SD)-induced elevations of c-jun and c-fos, but not NGFI-A, mRNA. fcl 17-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 11072104-8 2000 Prazosin reduced FCL(SD)-induced elevations of c-jun and c-fos, but not NGFI-A, mRNA. SD 0006 21-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 11072104-9 2000 Methoxyidazoxan enhanced NGFI-A and c-fos mRNA expression after FCL(SD), but reduced c-jun. methoxyidazoxan 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 11069127-0 2000 Distribution of c-fos mRNA in the brain following intracerebroventricular injection of nitric oxide (NO)-releasing compounds: possible role of NO in central cardiovascular regulation. Nitric Oxide 87-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 11078453-0 2000 Repeated 2-deoxy-D-glucose-induced glucoprivation attenuates Fos expression and glucoregulatory responses during subsequent glucoprivation. Deoxyglucose 9-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 11078453-11 2000 Double-label immunohistochemistry showed that Fos-ir was reduced or abolished in catecholamine cell groups A1, A1/C1, C1, C3, and A6 and in the paraventricular nucleus of the hypothalamus and adrenal medulla. Catecholamines 81-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 11078453-12 2000 In other brain sites, 2DG-induced Fos-ir was diminished or unaffected by prior glucoprivation. Deoxyglucose 22-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 11031079-6 2000 Five days later, rats were sacrificed within 2 h of amphetamine injection to examine amphetamine-induced Fos expression in the striatum, a measure of dopaminergic-dependent function in target neurons. Amphetamine 85-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-108 11069119-4 2000 The effects of ghrelin and GHRP-6 to induce Fos or Egr-1 protein expression was significantly greater in fasted than in fed rats. ghrp 27-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 11069127-8 2000 injection of L-NAME induced c-fos mRNA expression in PVN, SON, locus coeruleus and NTS, suggesting a tonic inhibition of neuronal activity by NO or stimulation of neuronal activity by endogenous NO. NG-Nitroarginine Methyl Ester 13-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 11070194-0 2000 Effects of repeated methyl levodopa administration on apomorphine sensitivity of rotational behavior and striatal Fos expression of rats with unilateral 6-OHDA lesions. Oxidopamine 153-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 11058452-6 2000 Fos expression increased in the nucleus of the tractus solitarius during parturition in rats pretreated with either vehicle or progesterone, but not in rats that had been pretreated with progesterone and induced with oxytocin, indicating that this input was inhibited. Progesterone 127-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 11058452-7 2000 Endogenous opioids inhibit oxytocin neurones in late pregnancy and the opioid antagonist, naloxone, increases Fos expression in supraoptic nuclei by preventing inhibition. Naloxone 90-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 11058452-8 2000 However, progesterone attenuated naloxone-induced Fos expression in the supraoptic nucleus in late pregnancy and naloxone administered during parturition did not accelerate the duration of births delayed by progesterone administration, indicating that progesterone does not act by hyperactivation of endogenous opioid tone. Progesterone 9-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 11058452-8 2000 However, progesterone attenuated naloxone-induced Fos expression in the supraoptic nucleus in late pregnancy and naloxone administered during parturition did not accelerate the duration of births delayed by progesterone administration, indicating that progesterone does not act by hyperactivation of endogenous opioid tone. Naloxone 33-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 11058452-9 2000 RU486, a progesterone receptor antagonist, enhanced supraoptic neurone Fos expression in late pregnancy, indicating progesterone receptor-mediated actions. Mifepristone 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 11124586-0 2000 Estrogen and tamoxifen differentially regulate beta-endorphin and cFos expression and neuronal colocalization in the arcuate nucleus of the rat. Tamoxifen 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-70 11124587-6 2000 Forty-eight-hour estradiol (10 microg) administration to OVX female rats increased AngII-induced c-Fos labeling in the lateral magnocellular neurons of the PVN by 30% as compared to vehicle-treated controls. Estradiol 17-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 11070194-7 2000 Repeated administration of methyl levodopa resulted in diminished apomorphine sensitivity of Fos expression in the dopamine depleted caudate-putamen and in enhanced sensitivity in the globus pallidus of the same side. methyl levodopa 27-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 11101146-5 2000 However, the induction of c-fos mRNA was detected as early as 5 min after dopamine treatment, peaked at 60 min, and remained elevated 5 h after treatment. Dopamine 74-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 11070194-7 2000 Repeated administration of methyl levodopa resulted in diminished apomorphine sensitivity of Fos expression in the dopamine depleted caudate-putamen and in enhanced sensitivity in the globus pallidus of the same side. Apomorphine 66-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 11101146-6 2000 Temporal profiles of increases in c-fos mRNA by R(+)-SKF-38393 (50 microM) and forskolin (50 microM) were similar to that of dopamine. Colforsin 79-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 11070194-7 2000 Repeated administration of methyl levodopa resulted in diminished apomorphine sensitivity of Fos expression in the dopamine depleted caudate-putamen and in enhanced sensitivity in the globus pallidus of the same side. Dopamine 115-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 11101146-9 2000 However, when cells were treated with dopamine, an increase in the expression of c-Fos immunoreactivity was observed after treatment for 2 h. The treatment of hippocampal neurons with R(+)-SKF38393 (50 microM) or forskolin (50 microM) also induced a significant increase in c-Fos expression. Dopamine 38-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 11101146-9 2000 However, when cells were treated with dopamine, an increase in the expression of c-Fos immunoreactivity was observed after treatment for 2 h. The treatment of hippocampal neurons with R(+)-SKF38393 (50 microM) or forskolin (50 microM) also induced a significant increase in c-Fos expression. Dopamine 38-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 274-279 11109985-8 2000 In contrast to the effects of oleate infusion, maltotriose infusion significantly and similarly increased Fos-li nuclei in the hindbrains of both LF and HF rats. maltotriose 47-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 11101146-9 2000 However, when cells were treated with dopamine, an increase in the expression of c-Fos immunoreactivity was observed after treatment for 2 h. The treatment of hippocampal neurons with R(+)-SKF38393 (50 microM) or forskolin (50 microM) also induced a significant increase in c-Fos expression. Colforsin 213-222 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 11101146-10 2000 These results indicate that the dopamine D1 receptor-mediated cAMP dependent pathway is associated with the expression of c-Fos in the hippocampal neurons. Cyclic AMP 62-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 11860940-0 2000 [Effects of fluoride on the expression of c-fos and c-jun genes and cell proliferation of rat osteoblasts]. Fluorides 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 11860940-1 2000 OBJECTIVE: To investigate the effects of sodium fluoride (NaF) on the expression of c-Fos and c-Jun genes and osteoblast cell proliferation of rat osteoblasts. Sodium Fluoride 41-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 11860940-1 2000 OBJECTIVE: To investigate the effects of sodium fluoride (NaF) on the expression of c-Fos and c-Jun genes and osteoblast cell proliferation of rat osteoblasts. Sodium Fluoride 58-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 12520955-0 2000 [Effects of cadmium on the expression of proto-oncogene c-fos and c-jun of liver cell in rats]. Cadmium 12-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-61 12520955-1 2000 The effects of cadmium chloride on the expression of proto-oncogene c-fos and c-jun of rat liver cells were studied with Northern dot hybridization. Cadmium Chloride 15-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-73 12520955-2 2000 The results showed that at the dose of 5, 10 or 20 mumol/kg of cadmium chloride, the expression of proto-oncogene c-fos and c-jun could be induced obviously. Cadmium Chloride 63-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-119 11860940-4 2000 RESULTS: RESULTS: from the MTT assay showed that NaF increased the proliferation of rat osteoblast and induced the expression of c-Fos and c-Jun genes. monooxyethylene trimethylolpropane tristearate 28-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 11860940-4 2000 RESULTS: RESULTS: from the MTT assay showed that NaF increased the proliferation of rat osteoblast and induced the expression of c-Fos and c-Jun genes. Sodium Fluoride 50-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 11860940-5 2000 The increases of c-Fos and c-Jun expression by the 3 different doses of NaF were 5.4%, 15.4% and 42.3% for the c-Fos gene and 12.1%, 14.4% and 38.6% for c-Jun gene respectively (P < 0.05 and P < 0.01). Sodium Fluoride 72-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 11860940-5 2000 The increases of c-Fos and c-Jun expression by the 3 different doses of NaF were 5.4%, 15.4% and 42.3% for the c-Fos gene and 12.1%, 14.4% and 38.6% for c-Jun gene respectively (P < 0.05 and P < 0.01). Sodium Fluoride 72-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 11106001-0 2000 The effect of the antipsychotic drug mosapramine on the expression of Fos protein in the rat brain: comparison with haloperidol, clozapine and risperidone. mosapramine 37-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 11106001-2 2000 administration of the antipsychotic drug mosapramine, as well as the antipsychotic drugs clozapine, haloperidol and risperidone, on the expression of Fos protein in the medial prefrontal cortex, nucleus accumbens and dorsolateral striatum of rat brain. mosapramine 41-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-153 11106001-2 2000 administration of the antipsychotic drug mosapramine, as well as the antipsychotic drugs clozapine, haloperidol and risperidone, on the expression of Fos protein in the medial prefrontal cortex, nucleus accumbens and dorsolateral striatum of rat brain. Clozapine 89-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-153 10992247-3 2000 In light of its widespread biological actions, immunohistochemical detection of the c-Fos protein product was used to study the distribution of neuronal activation in the rat brain caused by intraventricular (icv) injections of the selective NK(3) receptor agonist (succinyl-[Asp(6), N-Me-Phe(8)] substance P [6-11]), senktide. succinyl-[asp 266-279 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 11106001-2 2000 administration of the antipsychotic drug mosapramine, as well as the antipsychotic drugs clozapine, haloperidol and risperidone, on the expression of Fos protein in the medial prefrontal cortex, nucleus accumbens and dorsolateral striatum of rat brain. Haloperidol 100-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-153 11106001-2 2000 administration of the antipsychotic drug mosapramine, as well as the antipsychotic drugs clozapine, haloperidol and risperidone, on the expression of Fos protein in the medial prefrontal cortex, nucleus accumbens and dorsolateral striatum of rat brain. Risperidone 116-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-153 11106001-3 2000 The administration of mosapramine (1 or 3 mg/kg) significantly increased the number of Fos protein positive neurons in the medial prefrontal cortex, but not in the dorsolateral striatum. mosapramine 22-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 10992247-3 2000 In light of its widespread biological actions, immunohistochemical detection of the c-Fos protein product was used to study the distribution of neuronal activation in the rat brain caused by intraventricular (icv) injections of the selective NK(3) receptor agonist (succinyl-[Asp(6), N-Me-Phe(8)] substance P [6-11]), senktide. N-Methyl-L-phenylalanine 284-292 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 10992247-4 2000 Quantitative analysis revealed that treatment with isotonic saline or 200 ng senktide resulted in the differential expression of Fos-like immunoreactivity (FLI) throughout the brain. Sodium Chloride 60-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-132 11106001-4 2000 In addition, mosapramine (1, 3 or 10 mg/kg) produced a dose-dependent increase in the number of Fos protein positive neurons in the nucleus accumbens. mosapramine 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-99 11106001-5 2000 The acute administration of 10 mg/kg of mosapramine significantly increased the number of Fos protein positive neurons in all brain regions. mosapramine 40-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 11106001-6 2000 The acute administration of clozapine (30 mg/kg), similarly to mosapramine at lower doses (1 or 3 mg/kg), significantly increased the number of Fos protein positive neurons in the medial prefrontal cortex and nucleus accumbens, but not dorsolateral striatum. Clozapine 28-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-147 11106001-7 2000 In contrast, haloperidol (0.3 mg/kg) significantly increased the number of Fos protein positive neurons in the nucleus accumbens and dorsolateral striatum, but not medial prefrontal cortex. Haloperidol 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 11106001-9 2000 These results suggest that the ability of mosapramine to enhance expression of Fos protein in the medial prefrontal cortex may contribute to a clozapine-like profile with respect to actions on negative symptoms in schizophrenia. mosapramine 42-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 11106001-9 2000 These results suggest that the ability of mosapramine to enhance expression of Fos protein in the medial prefrontal cortex may contribute to a clozapine-like profile with respect to actions on negative symptoms in schizophrenia. Clozapine 143-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 11011010-7 2000 Furthermore, L-DOPA produced a dramatic induction of c-Fos in the CPu in 6-OHDA-lesioned animals. Levodopa 13-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 11011010-7 2000 Furthermore, L-DOPA produced a dramatic induction of c-Fos in the CPu in 6-OHDA-lesioned animals. Oxidopamine 73-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 11011034-0 2000 Effects of flurbiprofen and its enantiomers on the spinal c-Fos protein expression induced by noxious heat stimuli in the anaesthetized rat. Flurbiprofen 11-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 11501038-11 2000 CONCLUSION: In the 6-OHDA-lesioned rats, the rotation induced by SPD was kept on a high activity, which was in pace with the inducement of Fos expression and the reduction of expression of PENK mRNA in the denervated striatum. Oxidopamine 19-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 11011034-1 2000 We have evaluated the effects of either intravenous or intraplantar administration of racemic-, S(+)- and R(-)-flurbiprofen on the spinal c-Fos protein expression after a single noxious heat stimulation (52 degrees C for 15 s) of the rat hindpaw in urethane anaesthetized rats. -, s(+)- and r(-)-flurbiprofen 93-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 11011034-4 2000 The same doses of R(-)-flurbiprofen produced dose-related effects (r=0.58, P<0.05) with weak, but significant, effects for doses of 3 and 9 mg/kg (18+/-6% and 26+/-5% reduction of the number of noxious heat-evoked c-Fos-IR nuclei in laminae I-II, P<0.05 and P<0.01, respectively). Flurbiprofen 18-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 217-222 11009450-7 2000 LIF-induced expression of c-fos, brain natriuretic peptide, and skeletal alpha-actin mRNA was markedly suppressed by PD-98059 and moderately suppressed by wortmannin and AG-490. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 117-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 11009450-7 2000 LIF-induced expression of c-fos, brain natriuretic peptide, and skeletal alpha-actin mRNA was markedly suppressed by PD-98059 and moderately suppressed by wortmannin and AG-490. Wortmannin 155-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 11173339-7 2000 This finding partly differs from previous studies where Fos expression occurred by other reasons such as hypovolemia, hemorrhage, nitroprusside or hydralazine etc. Nitroprusside 130-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 11173339-7 2000 This finding partly differs from previous studies where Fos expression occurred by other reasons such as hypovolemia, hemorrhage, nitroprusside or hydralazine etc. Hydralazine 147-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 11069385-4 2000 Acetic acid was instilled into the bladder to induce c-fos expression. Acetic Acid 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 11057525-8 2000 Subsequent testing with DMCM confirmed that a benzodiazepine inverse agonist can induce Fos-LI in most of the same brain regions as observed following ethanol withdrawal, and that this change in Fos protein can be attenuated by pretreatment with flumazenil (5.0 mg/kg). Flumazenil 246-256 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-198 11501038-1 2000 AIM: To study that l-stepholidine (SPD) regulates the expression of proenkephalin (PENK) and prodynorphin (PDYN) mRNA and Fos in the striatum after rotational test in the 6-hydroxydopamine (6-OHDA)-lesioned rats. stepholidine 19-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 11501038-1 2000 AIM: To study that l-stepholidine (SPD) regulates the expression of proenkephalin (PENK) and prodynorphin (PDYN) mRNA and Fos in the striatum after rotational test in the 6-hydroxydopamine (6-OHDA)-lesioned rats. stepholidine 35-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 11044751-0 2000 Nicotine-induced fos expression in the pedunculopontine mesencephalic tegmentum in the rat. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 11044751-1 2000 The aim of this study was to assess the effects of a single dose of nicotine (NIC, 0.3 or 1.0 mg/kg, s.c.), after survival times of 30, 60 or 120 min, on immediate early gene expression in the pedunculopontine mesencephalic tegmentum (PMT), using Fos-immunocytochemistry. Nicotine 68-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 247-250 11057524-3 2000 Recent studies point to central adaptive changes insofar as rewarding, locomotor and c-fos-inducing effects of amphetamine and MK-801, injected directly into the lateral ventricle, are greater in food-restricted than ad libitum fed rats. Amphetamine 111-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 11057524-3 2000 Recent studies point to central adaptive changes insofar as rewarding, locomotor and c-fos-inducing effects of amphetamine and MK-801, injected directly into the lateral ventricle, are greater in food-restricted than ad libitum fed rats. Dizocilpine Maleate 127-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 11057524-4 2000 The increased expression of c-fos in nucleus accumbens (NAC) shell, in particular, suggests that food restriction may augment drug reward by modulating dopamine (DA) synaptic function in this area. Dopamine 152-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 11057524-4 2000 The increased expression of c-fos in nucleus accumbens (NAC) shell, in particular, suggests that food restriction may augment drug reward by modulating dopamine (DA) synaptic function in this area. Dopamine 162-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 11057525-5 2000 In addition, flumazenil effects on Fos-LI were measured in a group of animals treated with the BZD inverse agonist DMCM (0.75 and 1.0 mg/kg). Flumazenil 13-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 11057525-8 2000 Subsequent testing with DMCM confirmed that a benzodiazepine inverse agonist can induce Fos-LI in most of the same brain regions as observed following ethanol withdrawal, and that this change in Fos protein can be attenuated by pretreatment with flumazenil (5.0 mg/kg). methyl 6,7-dimethoxy-4-ethyl-beta-carboline-3-carboxylate 24-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-198 11057525-8 2000 Subsequent testing with DMCM confirmed that a benzodiazepine inverse agonist can induce Fos-LI in most of the same brain regions as observed following ethanol withdrawal, and that this change in Fos protein can be attenuated by pretreatment with flumazenil (5.0 mg/kg). Benzodiazepines 46-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 11057525-8 2000 Subsequent testing with DMCM confirmed that a benzodiazepine inverse agonist can induce Fos-LI in most of the same brain regions as observed following ethanol withdrawal, and that this change in Fos protein can be attenuated by pretreatment with flumazenil (5.0 mg/kg). Benzodiazepines 46-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-198 11057525-8 2000 Subsequent testing with DMCM confirmed that a benzodiazepine inverse agonist can induce Fos-LI in most of the same brain regions as observed following ethanol withdrawal, and that this change in Fos protein can be attenuated by pretreatment with flumazenil (5.0 mg/kg). Ethanol 151-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-198 11057525-8 2000 Subsequent testing with DMCM confirmed that a benzodiazepine inverse agonist can induce Fos-LI in most of the same brain regions as observed following ethanol withdrawal, and that this change in Fos protein can be attenuated by pretreatment with flumazenil (5.0 mg/kg). Flumazenil 246-256 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 10996146-7 2000 CCK-induced Fos expression was abolished or attenuated in the brains of vagotomized or capsaicin-treated animals. Capsaicin 87-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-15 11043526-2 2000 Subcutaneous nitroglycerin (10 mg/kg) produced a significant increase of nitric oxide synthase (NOS)- and c-fos-immunoreactive neurons in the cervical part of trigeminal nucleus caudalis in rats after 4 h. This effect was not observed in the thoracic dorsal horn. Nitroglycerin 13-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 11043526-3 2000 Similar increase of NOS and c-fos was obtained in the brain stem after a somatic nociceptive stimulus, i.e. on the side of the formalin injection in the lip. Formaldehyde 127-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 10984628-5 2000 Double fluorescence immunohistochemistry for c-Fos and serotonin revealed that PAG/NRD/EW neurons expressing c-Fos were non-serotonergic. Serotonin 55-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 10986339-3 2000 Changes in neuronal activity were assessed by immunohistochemical analysis of expression of an inducible transcription factor (ITF), c-Fos, in the brain of rats habituated to repeated pentobarbital anesthesia or saline administration. Pentobarbital 184-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 10986339-10 2000 On the other hand, pentobarbital by itself strongly induced c-Fos expression in the lateral habenula of saline-, cocaine-, and ethanol-injected rats. Pentobarbital 19-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 10986339-10 2000 On the other hand, pentobarbital by itself strongly induced c-Fos expression in the lateral habenula of saline-, cocaine-, and ethanol-injected rats. Sodium Chloride 104-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 10986339-10 2000 On the other hand, pentobarbital by itself strongly induced c-Fos expression in the lateral habenula of saline-, cocaine-, and ethanol-injected rats. Cocaine 113-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 10986339-10 2000 On the other hand, pentobarbital by itself strongly induced c-Fos expression in the lateral habenula of saline-, cocaine-, and ethanol-injected rats. Ethanol 127-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 10986349-0 2000 Differential Fos expression in the paraventricular nucleus of the hypothalamus, sacral parasympathetic nucleus and colonic motor response to water avoidance stress in Fischer and Lewis rats. Water 141-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 10986349-3 2000 Exposure for 60 min to water avoidance stress increased the number of Fos positive cells in the paraventricular nucleus of the hypothalamus (PVN), nucleus tractus solitarius (NTS), and the parasympathetic nucleus of the lumbo-sacral spinal cord (L6-S1) in both Lewis and Fischer rats compared with non stress groups. Water 23-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 10973588-0 2000 Neuronal Fos-like immunoreactivity in ouabain-induced hypertension. Ouabain 38-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 10973597-3 2000 Intracerebroventricular administration of N/OFQ induces changes in c-Fos immunoreactivity in several feeding-related brain sites. Nitrogen 42-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 10973597-6 2000 We also evaluated c-Fos immunoreactivity in those areas of the brain which have been shown to exhibit altered c-Fos expression upon N/OFQ administration. Nitrogen 132-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 11085307-0 2000 C-fos and egr-1 immediate-early gene induction by cocaine and cocaethylene in rat brain: a comparative study. Cocaine 50-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 11085307-0 2000 C-fos and egr-1 immediate-early gene induction by cocaine and cocaethylene in rat brain: a comparative study. cocaethylene 62-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 11085307-4 2000 cocaine (20 mg/kg) injection produced a strong expression of egr-1 and c-fos genes in the nucleus accumbens, caudate-putamen, and frontal cortex in the rat. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 11085307-7 2000 A high dose of ethanol increased egr-1 and c-fos expression in the frontal cortex and in the lateral part of the caudate-putamen. Ethanol 15-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 11085309-0 2000 Microtubule-associated protein 2 (MAP2) and c-fos expression in the rat prefrontal cortex following subchronic treatment with substituted amphetamines. Amphetamines 138-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 11023626-9 2000 Animals receiving a serotonin subtype three (5-HT3, 5-hydroxytryptamine) receptor antagonist (tropisetron, ICS 205-930, 3-tropanyl-indole-3-carboxylate) showed a significant reduction in c-Fos protein expression compared to animals receiving a vehicle. 3-tropanyl-indole-3-carboxylate 120-151 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 187-192 11124386-5 2000 Eighteen hours later, rats were injected with either saline or 2-deoxy glucose (2-DG) and brain sections were stained to demonstrate 2-DG-activated neurons immunoreactive for Fos protein. Deoxyglucose 133-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 175-178 10993216-3 2000 After the administration of 3 mg/kg ketanserin, the expression of Fos-ir cells in the periglomerular cell layer in response to male Wistar urine was inhibited, while that in the mitral/tufted cell and granule cell layers was not changed. Ketanserin 36-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 10993216-4 2000 The administration of 20 mg/kg propranolol inhibited the expression of Fos-ir cells in all three layers. Propranolol 31-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 10993216-5 2000 These results suggest that serotonin and noradrenaline are likely involved in the modulation of the expression of Fos-ir cells in response to the urine in the accessory olfactory bulb. Serotonin 27-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 10993216-5 2000 These results suggest that serotonin and noradrenaline are likely involved in the modulation of the expression of Fos-ir cells in response to the urine in the accessory olfactory bulb. Norepinephrine 41-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 10967299-0 2000 Expression of estrogen receptor-alpha and c-Fos in adrenergic neurons of the female rat during the steroid-induced LH surge. Steroids 99-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 10967299-0 2000 Expression of estrogen receptor-alpha and c-Fos in adrenergic neurons of the female rat during the steroid-induced LH surge. Luteinizing Hormone 115-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 10998122-1 2000 The c-fos immediate-early gene is induced by morphine and other drugs of abuse in the nucleus accumbens (NAc), a mesolimbic region implicated in drug abuse and reward. Morphine 45-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 10998122-3 2000 CPP was completely prevented by c-fos antisense ODN infused bilaterally into the NAc prior to each systemic morphine injection, whereas sense and missense NAc injections had no effect on CPP. Morphine 108-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 11068700-1 2000 Dietary short-chain fructooligosaccharides (Sc-FOS) increase mucosal calbindin-D9k (CaBP) levels in the large intestine whereas levels in the small intestine are decreased in rats. fructooligosaccharide 20-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 11068700-2 2000 In the present study, we investigated the mechanism by which Sc-FOS induce this increase in CaBP in the large intestine by measuring intestinal CaBP levels in rats fed normal and calcium-deficient diets. Calcium 179-186 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 10963887-5 2000 It was revealed that intramuscular injection of formalin, in the anaesthetised rat, evoked a significant increase in Fos expression within the caudal vlPAG, and that approximately 25% of the Fos-immunoreactive neurons projected to the RVLM. Formaldehyde 48-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 10963887-5 2000 It was revealed that intramuscular injection of formalin, in the anaesthetised rat, evoked a significant increase in Fos expression within the caudal vlPAG, and that approximately 25% of the Fos-immunoreactive neurons projected to the RVLM. Formaldehyde 48-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 191-194 10963888-0 2000 Intracerebroventricular administration of the beta(3)-adrenoceptor agonist CL 316243 causes Fos immunoreactivity in discrete regions of rat hypothalamus. disodium (R,R)-5-(2-((2-(3-chlorophenyl)-2-hydroxyethyl)-amino)propyl)-1,3-benzodioxole-2,3-dicarboxylate 75-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 10963888-4 2000 administration of the selective beta(3)-AR agonist CL316243 on Fos expression to determine whether beta(3)-AR stimulation affects neurones within specific brain nuclei. disodium (R,R)-5-(2-((2-(3-chlorophenyl)-2-hydroxyethyl)-amino)propyl)-1,3-benzodioxole-2,3-dicarboxylate 51-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 10963888-7 2000 Pre-treatment with the selective beta(3)-AR antagonist SR59230A resulted in a significant decrease in the number of Fos positive cells in all those areas compared with rats treated with CL316243 alone. 3-(2-ethylphenoxy)-1-(1,2,3,4-tetrahydronaphth-1-ylamino)-2-propanol oxalate 55-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 10966845-7 2000 GAL4-Elk1 studies revealed that DEX suppressed TGF beta-induced ERK activation which led to c-fos gene expression followed by increase in AP-1 complex formation, whereas the Smad pathway was not involved in DEX-dependent negative regulation of AP-1 in a reporter assay that requires FAST1-Smad2 for the activation. Dexamethasone 32-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 10966845-8 2000 DEX also eliminated TGF beta-induced c-fos mRNA expression and ERK activation in Northern analysis and in vitro kinase assay, respectively. Dexamethasone 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 11026743-5 2000 Furthermore, SKF 82958 produced a dramatic induction of c-Fos in the DR, an effect that was blocked by 8-OH-DPAT. 8-Hydroxy-2-(di-n-propylamino)tetralin 103-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 10940358-1 2000 Involvement of c-fos and neuronal nitric oxide synthase (nNOS) in the hyperalgesia induced by complete Freund adjuvant (CFA) has been reported. complete freund adjuvant 94-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 10940358-1 2000 Involvement of c-fos and neuronal nitric oxide synthase (nNOS) in the hyperalgesia induced by complete Freund adjuvant (CFA) has been reported. 3-chloro-4-fluoroaniline 120-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 10880996-3 2000 Subcutaneous capsaicin injection in male rats, compared with vehicle, caused a significant increase in Fos expression in the paraventricular nucleus (PVN), supraoptic nucleus (SON), and medial and cortical amygdala. Capsaicin 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-106 10880996-6 2000 A higher proportion of the NOS neurons in the PVN, periventricular region, SON and amygdala showed Fos expression in response to capsaicin than vehicle injection. Capsaicin 129-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 10962128-3 2000 The results show that hydroxyl radicals generated by the Fenton reaction induced apoptosis in cerebellar granule cells, which was associated with the decrease in the Bcl-2 mRNA level and the increase in the protein levels of the transcription factors Fos and Jun. Hydroxyl Radical 22-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 251-254 10904129-0 2000 A comparison of the patterns of striatal Fos-like immunoreactivity induced by various dopamine agonists in rats. Dopamine 86-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 10904129-1 2000 In contrast to the highly patchy patterns of Fos-like immunoreactivity seen in the rostral striatum after administration of a number of dopamine agonists, the monoamine uptake blocker cocaine has been reported to produce a relatively homogeneous pattern of gene expression. Dopamine 136-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 10904129-2 2000 In the current study we extended these observations by using a quantitative technique to demonstrate that while amphetamine and apomorphine produce patchy striatal Fos expression, the selective dopamine uptake inhibitors amfonelic acid, nomifensine and GBR-12909 all, like cocaine, produce near random patterns of gene expression. Amphetamine 112-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-167 10904129-2 2000 In the current study we extended these observations by using a quantitative technique to demonstrate that while amphetamine and apomorphine produce patchy striatal Fos expression, the selective dopamine uptake inhibitors amfonelic acid, nomifensine and GBR-12909 all, like cocaine, produce near random patterns of gene expression. Apomorphine 128-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-167 10943690-0 2000 Differential expression of c-fos following administration of two tremorgenic agents: harmaline and oxotremorine. Harmaline 85-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 10943690-0 2000 Differential expression of c-fos following administration of two tremorgenic agents: harmaline and oxotremorine. Oxotremorine 99-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 10943690-2 2000 In both the harmaline- and oxotremorin-treated rats, c-Fos-positive neurons were extensively distributed in the basal ganglia nuclei and the cerebellum. Harmaline 12-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 10943690-2 2000 In both the harmaline- and oxotremorin-treated rats, c-Fos-positive neurons were extensively distributed in the basal ganglia nuclei and the cerebellum. Oxotremorine 27-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 10943690-3 2000 Additionally, in the harmaline-treated rats, numerous c-Fos-positive neurons were also distributed throughout the inferior olivary nucleus. Harmaline 21-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 10943690-4 2000 In the oxotremorine-treated rats, while the inferior olive was not involved, c-Fos was strongly expressed in the neurons of the reticular thalamic nucleus, possibly due to the muscarinic effects of oxotremorine. Oxotremorine 7-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 10943690-4 2000 In the oxotremorine-treated rats, while the inferior olive was not involved, c-Fos was strongly expressed in the neurons of the reticular thalamic nucleus, possibly due to the muscarinic effects of oxotremorine. Oxotremorine 198-210 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 10967546-5 2000 While c-fos was induced in UMR cells, both c-fos and jun B were induced in ROS cells. ros 75-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 10840165-3 2000 In addition, onapristone induces expression of c-fos mRNA, which is induced by estrogens but not progestins in this target tissue. onapristone 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 10840165-4 2000 This induction of VEGF and c-fos by onapristone is inhibited by the antiestrogen ICI 182,780, but not by the antiprogestin RU-486. onapristone 36-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 10919270-0 2000 Estradiol stimulation of c-fos and c-jun expressions and activator protein-1 deoxyribonucleic acid binding activity in rat white adipocyte. Estradiol 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 10919270-1 2000 In order to elucidate the molecular mechanisms whereby ovarian hormones, and particularly estrogens, modulate fat cell metabolism, we investigated the effects of estradiol administration on c-fos and c-jun expressions in fat cells from ovariectomized (OVX) rats. Estradiol 162-171 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 190-195 10919270-2 2000 Estradiol treatment resulted in a rapid increase in c-fos and c-jun messenger RNA (mRNA) and protein levels (about 2-fold). Estradiol 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 10919270-3 2000 These effects of estradiol on c-fos and c-jun mRNAs were blocked by actinomycin D but not by cycloheximide treatment, suggesting that estradiol modulates c-fos and c-jun transcription. Estradiol 17-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 10919270-3 2000 These effects of estradiol on c-fos and c-jun mRNAs were blocked by actinomycin D but not by cycloheximide treatment, suggesting that estradiol modulates c-fos and c-jun transcription. Estradiol 17-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 10919270-3 2000 These effects of estradiol on c-fos and c-jun mRNAs were blocked by actinomycin D but not by cycloheximide treatment, suggesting that estradiol modulates c-fos and c-jun transcription. Dactinomycin 68-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 10919270-3 2000 These effects of estradiol on c-fos and c-jun mRNAs were blocked by actinomycin D but not by cycloheximide treatment, suggesting that estradiol modulates c-fos and c-jun transcription. Dactinomycin 68-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 10919270-3 2000 These effects of estradiol on c-fos and c-jun mRNAs were blocked by actinomycin D but not by cycloheximide treatment, suggesting that estradiol modulates c-fos and c-jun transcription. Estradiol 134-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 10919270-3 2000 These effects of estradiol on c-fos and c-jun mRNAs were blocked by actinomycin D but not by cycloheximide treatment, suggesting that estradiol modulates c-fos and c-jun transcription. Estradiol 134-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 10919270-7 2000 In addition, the estradiol-induced changes in c-fos and c-jun mRNA expressions could not be observed in castrated males suggesting a gender-specific effect of estradiol. Estradiol 17-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 10919270-7 2000 In addition, the estradiol-induced changes in c-fos and c-jun mRNA expressions could not be observed in castrated males suggesting a gender-specific effect of estradiol. Estradiol 159-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 10985689-0 2000 Caudal brainstem Fos expression is restricted to periventricular catecholamine neuron-containing loci following intraventricular administration of 2-deoxy-D-glucose. Catecholamines 65-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 10985689-0 2000 Caudal brainstem Fos expression is restricted to periventricular catecholamine neuron-containing loci following intraventricular administration of 2-deoxy-D-glucose. Deoxyglucose 147-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 10985689-2 2000 In the present study, Fos immunocytochemistry was employed to characterize the distribution of neurons within this region of the male rat brain that undergo genomic activation in response to intraventricular delivery of the antiglycolytic agent, 2-deoxy-D-glucose (2DG). Deoxyglucose 246-263 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-25 10915578-6 2000 The significant induction of c-fos mRNA by audiogenic seizures in PTU rats or by AMPA- or cyclothiazide-induced seizures in naive rats was prominent in the IC. cyclothiazide 90-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 10915578-7 2000 MK-801 suppressed c-fos mRNA expression in the IC induced by audiogenic seizures in PTU rats or by AMPA-induced seizures in naive rats. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 10915578-8 2000 NBQX suppressed the expression of c-fos mRNA in the IC induced by AMPA-induced seizures but did not suppress c-fos mRNA in PTU rats or rats with cyclothiazide-induced seizures. 2,3-dioxo-6-nitro-7-sulfamoylbenzo(f)quinoxaline 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 10915578-8 2000 NBQX suppressed the expression of c-fos mRNA in the IC induced by AMPA-induced seizures but did not suppress c-fos mRNA in PTU rats or rats with cyclothiazide-induced seizures. alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid 66-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 10929091-7 2000 Therefore, to obtain some indication of oestrogen"s possible site(s) of action, Fos-like immunolabelling was mapped in the amygdala and in brainstem catecholamine groups, which are neuronal populations demonstrating substantial evidence of involvement in the generation of HPA axis stress responses. Catecholamines 149-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 10883804-6 2000 Because the conditions under which Fos-IR expression occurred in females are identical to those required for sperm transport, we suggest that, in the female, sperm transport is regulated in part by autonomic outflow from the PG after copulation. pg 225-227 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 10882842-0 2000 Clozapine- and olanzapine-induced Fos expression in the rat medial prefrontal cortex is mediated by beta-adrenoceptors. Clozapine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 10882842-0 2000 Clozapine- and olanzapine-induced Fos expression in the rat medial prefrontal cortex is mediated by beta-adrenoceptors. Olanzapine 15-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 10882842-2 2000 Recently, it has been suggested that the ability of clozapine and olanzapine to induce Fos expression in the medial prefrontal cortex (mPFC), contribute to their therapeutic efficacy. Clozapine 52-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 10882842-2 2000 Recently, it has been suggested that the ability of clozapine and olanzapine to induce Fos expression in the medial prefrontal cortex (mPFC), contribute to their therapeutic efficacy. Olanzapine 66-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 10882842-4 2000 In the present study, we demonstrate that clozapine- and olanzapine-induced Fos expression in the mPFC are inhibited by propranolol. Clozapine 42-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 10882842-4 2000 In the present study, we demonstrate that clozapine- and olanzapine-induced Fos expression in the mPFC are inhibited by propranolol. Olanzapine 57-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 10882842-4 2000 In the present study, we demonstrate that clozapine- and olanzapine-induced Fos expression in the mPFC are inhibited by propranolol. Propranolol 120-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 10882842-5 2000 We also show that clozapine and olanzapine induce Fos expression in the locus coeruleus. Clozapine 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 10882842-5 2000 We also show that clozapine and olanzapine induce Fos expression in the locus coeruleus. Olanzapine 32-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 10882842-6 2000 These results suggest that clozapine and olanzapine increase noradrenaline release by stimulating noradrenergic neuronal activity in the locus coeruleus and, consequently, increased noradrenaline induce Fos expression in the mPFC via beta-adrenergic receptors. Clozapine 27-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 203-206 10882842-6 2000 These results suggest that clozapine and olanzapine increase noradrenaline release by stimulating noradrenergic neuronal activity in the locus coeruleus and, consequently, increased noradrenaline induce Fos expression in the mPFC via beta-adrenergic receptors. Olanzapine 41-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 203-206 10882842-6 2000 These results suggest that clozapine and olanzapine increase noradrenaline release by stimulating noradrenergic neuronal activity in the locus coeruleus and, consequently, increased noradrenaline induce Fos expression in the mPFC via beta-adrenergic receptors. Norepinephrine 182-195 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 203-206 12749994-0 2003 The effects of dexmedetomidine and halothane on Fos expression in the spinal dorsal horn using a rat postoperative pain model. Dexmedetomidine 15-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 12749994-0 2003 The effects of dexmedetomidine and halothane on Fos expression in the spinal dorsal horn using a rat postoperative pain model. Halothane 35-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 12749994-1 2003 We investigated the effect of an intrathecal injection of a selective alpha2 adrenergic receptor agonist, dexmedetomidine (Dex), and halothane anesthesia on Fos expression in the lumbar spinal dorsal horn after skin incision of the plantar surface of the hind paw, a postoperative pain model using rats. Dexmedetomidine 106-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-160 12749994-1 2003 We investigated the effect of an intrathecal injection of a selective alpha2 adrenergic receptor agonist, dexmedetomidine (Dex), and halothane anesthesia on Fos expression in the lumbar spinal dorsal horn after skin incision of the plantar surface of the hind paw, a postoperative pain model using rats. Dexmedetomidine 123-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-160 12749994-1 2003 We investigated the effect of an intrathecal injection of a selective alpha2 adrenergic receptor agonist, dexmedetomidine (Dex), and halothane anesthesia on Fos expression in the lumbar spinal dorsal horn after skin incision of the plantar surface of the hind paw, a postoperative pain model using rats. Halothane 133-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-160 12749994-3 2003 Halothane anesthesia (0.5-1.5%) partially reversed Fos induction, but not in a dose-dependent manner. Halothane 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 12749994-4 2003 Preoperative spinal Dex (0.1-10 microg) dose-dependently reduced Fos immunoreactivity, while a relatively high dose of Dex (10 microg) was necessary to produce a profound effect. Dexmedetomidine 20-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 12749994-5 2003 When used with halothane anesthesia, relatively low doses of Dex (1-3 microg) completely suppressed Fos induction in the superficial spinal layers. Dexmedetomidine 61-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-103 10976458-8 2000 In STZ-induced diabetic rats, Fos expression was also decreased by 300 lux of light. Streptozocin 3-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 10915909-0 2000 Maturation of locomotor and Fos responses to the NMDA antagonists, PCP and MK-801. N-Methylaspartate 49-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 10915909-0 2000 Maturation of locomotor and Fos responses to the NMDA antagonists, PCP and MK-801. Phencyclidine 67-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 10915909-0 2000 Maturation of locomotor and Fos responses to the NMDA antagonists, PCP and MK-801. Dizocilpine Maleate 75-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 10915909-6 2000 The age-dependent response to PCP may be related to developmental events in the motor cortex, since more Fos-immunoreactive neurons were observed in the motor cortex of prepubertal animals after PCP administration relative to adult animals. Phencyclidine 30-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-108 10915909-6 2000 The age-dependent response to PCP may be related to developmental events in the motor cortex, since more Fos-immunoreactive neurons were observed in the motor cortex of prepubertal animals after PCP administration relative to adult animals. Phencyclidine 195-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-108 10924699-0 2000 Regulation of c-fos gene expression by lipopolysaccharide and cycloheximide in C6 rat glioma cells. Cycloheximide 62-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 10924699-4 2000 Cycloheximide (CHX, 20 microM), a protein synthesis inhibitor, alone caused increases of c-fos and c-jun mRNA levels. Cycloheximide 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 10924699-4 2000 Cycloheximide (CHX, 20 microM), a protein synthesis inhibitor, alone caused increases of c-fos and c-jun mRNA levels. Cycloheximide 15-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 10899405-4 2000 Fos-expression was decreased following pretreatment with pyrilamine and ondansetron i.p. Pyrilamine 57-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 10899405-4 2000 Fos-expression was decreased following pretreatment with pyrilamine and ondansetron i.p. Ondansetron 72-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 10889339-3 2000 Noxious stimulation (subcutaneous injection of formalin into perioral regions) induced Fos-IR in some of GABA- and Gly-ir neurons. Formaldehyde 47-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 10889339-3 2000 Noxious stimulation (subcutaneous injection of formalin into perioral regions) induced Fos-IR in some of GABA- and Gly-ir neurons. gamma-Aminobutyric Acid 105-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 10889339-3 2000 Noxious stimulation (subcutaneous injection of formalin into perioral regions) induced Fos-IR in some of GABA- and Gly-ir neurons. Glycine 115-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 10908728-0 2000 Annexin-I inhibits PMA-induced c-fos SRE activation by suppressing cytosolic phospholipase A2 signal. Tetradecanoylphorbol Acetate 19-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 10908728-6 2000 The stimulation of Rat2 fibroblast cells with phorbol 12-myristate 13-acetate (PMA) induced the c-fos serum response element (SRE). Tetradecanoylphorbol Acetate 46-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 10908728-6 2000 The stimulation of Rat2 fibroblast cells with phorbol 12-myristate 13-acetate (PMA) induced the c-fos serum response element (SRE). Tetradecanoylphorbol Acetate 79-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 10896894-2 2000 In urethan-anesthetized Wistar rats after SCI for 6 wk, intravesical saline distension significantly (P </= 0.005) increased the number of Fos-immunoreactive (IR) cells in the rostrolumbar (L1, 38 cells/section; L2, 29 cells/section) and caudal lumbosacral (L6, 140 cells/section; S1, 110 cells/section) spinal cord compared with control animals, but Fos expression in the L5 segment was not altered. Urethane 3-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-145 10896894-2 2000 In urethan-anesthetized Wistar rats after SCI for 6 wk, intravesical saline distension significantly (P </= 0.005) increased the number of Fos-immunoreactive (IR) cells in the rostrolumbar (L1, 38 cells/section; L2, 29 cells/section) and caudal lumbosacral (L6, 140 cells/section; S1, 110 cells/section) spinal cord compared with control animals, but Fos expression in the L5 segment was not altered. Sodium Chloride 69-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-145 10896894-2 2000 In urethan-anesthetized Wistar rats after SCI for 6 wk, intravesical saline distension significantly (P </= 0.005) increased the number of Fos-immunoreactive (IR) cells in the rostrolumbar (L1, 38 cells/section; L2, 29 cells/section) and caudal lumbosacral (L6, 140 cells/section; S1, 110 cells/section) spinal cord compared with control animals, but Fos expression in the L5 segment was not altered. Sodium Chloride 69-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 354-357 10896894-8 2000 Pretreatment with capsaicin significantly reduced the number of Fos-IR cells induced by bladder distension after SCI. Capsaicin 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 10896894-9 2000 These data suggest that SCI can reveal an altered Fos expression pattern in response to a nonnoxious bladder stimulus that is partially mediated by capsaicin-sensitive bladder afferents. Capsaicin 148-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 10951896-5 2000 Furthermore, systemic injection of MK-801, a non-competitive antagonist of the NMDA receptor, reduced the LN-induced c-Fos expression in BPN and RtTg. Dizocilpine Maleate 35-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 10930210-8 2000 Sustained noxious stimulation to the hindpaw in halothane-anesthetized animals was associated with an increase in c-fos immunoreactivity in the dorsal horn of the lumbar spinal cord ipsilateral to the stimulation (P < 0.001). Halothane 48-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 10864974-0 2000 Nigrostriatal lesions alter oral dyskinesia and c-Fos expression induced by the serotonin agonist 1-(m-chlorophenyl)piperazine in adult rats. Serotonin 80-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 10864974-0 2000 Nigrostriatal lesions alter oral dyskinesia and c-Fos expression induced by the serotonin agonist 1-(m-chlorophenyl)piperazine in adult rats. 1-(3-chlorophenyl)piperazine 98-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 10864974-4 2000 Specifically, the effects of unilateral lesions of nigrostriatal DA neurons on oral dyskinesia and Fos protein expression induced by the non-selective 5-HT(2C) agonist 1-(m-chlorophenyl)piperazine (m-CPP) were examined. 1-(3-chlorophenyl)piperazine 168-196 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 10958117-2 2000 OBJECTIVE: To identify specific changes in monoamine and glutamate function in the nucleus accumbens and c-fos induction in the amygdala and striatum which may be correlated with altered cocaine self-administration in isolates. Cocaine 187-194 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 10958117-7 2000 Cocaine increased FOS expression in most amygdala and striatal brain regions examined that were relatively greater in isolation-reared rats in core and shell regions of the nucleus accumbens, medial and lateral regions of the dorsal striatum as well as the central nucleus of the amygdala. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 10901171-3 2000 The elevated c-fos mRNA level induced by CTX was significantly inhibited by the co-treatment with dexamethasone (DEX). Dexamethasone 98-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 10901171-3 2000 The elevated c-fos mRNA level induced by CTX was significantly inhibited by the co-treatment with dexamethasone (DEX). Dexamethasone 113-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 10901171-5 2000 Cycloheximide (CHX) increased c-fos and c-jun mRNA levels. Cycloheximide 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 10901171-5 2000 Cycloheximide (CHX) increased c-fos and c-jun mRNA levels. Cycloheximide 15-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 10901171-6 2000 CHX caused a super-induction of CTX-induced c-fos mRNA level. Cycloheximide 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 10901171-8 2000 In addition, DEX appears to have a selective inhibitory action against c-fos mRNA expression regulated by CTX. Dexamethasone 13-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 10819909-0 2000 Nicotine-induced Fos expression in the nucleus accumbens and the medial prefrontal cortex of the rat: role of nicotinic and NMDA receptors in the ventral tegmental area. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 10819909-2 2000 In the present study, we investigated the role of nicotinic and NMDA receptors in the VTA for the expression of Fos-like immunoreactivity (FLI) in the shell and core of the nucleus accumbens and in the medial prefrontal cortex (mPFC) of the rat after acute nicotine administration. Nicotine 257-265 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 10837795-6 2000 In addition, expression of p53 showed region- and gender-selective alterations consistent with cell damage; c-fos, which is constitutively overexpressed after gestational nicotine exposure, was unaffected with the adolescent treatment paradigm. Nicotine 171-179 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 10837802-1 2000 The object of the present study was to determine the c-fos gene expression pattern in the hypothalamus (HYP) and the preoptic area (POA) after estradiol and testosterone priming during the critical period of sexual differentiation of the rat brain. Testosterone 157-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 10837802-6 2000 We observed that c-fos gene expression was markedly increased in POA of the animals treated with estradiol or testosterone 2 h after treatments, while a non-significant increase in c-fos gene expression was observed in the HYP of these animals. Estradiol 97-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 10837802-6 2000 We observed that c-fos gene expression was markedly increased in POA of the animals treated with estradiol or testosterone 2 h after treatments, while a non-significant increase in c-fos gene expression was observed in the HYP of these animals. Testosterone 110-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 10837802-7 2000 We found a significant increase in c-fos expression in HYP and POA on the day of VO in both estradiol and testosterone defeminized rats. Vanadium(II) oxide 81-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 10837802-7 2000 We found a significant increase in c-fos expression in HYP and POA on the day of VO in both estradiol and testosterone defeminized rats. Testosterone 106-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 10837802-9 2000 The overall results suggest that estradiol and testosterone imprinting during critical postnatal period of sexual differentiation of the brain permanently modifies the regulation of c-fos gene expression. Testosterone 47-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 182-187 10856442-0 2000 Effects of antipsychotic drugs on neurotoxicity, expression of fos-like protein and c-fos mRNA in the retrosplenial cortex after administration of dizocilpine. Dizocilpine Maleate 147-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 10856442-1 2000 In this study, we examined the effect of clozapine, olanzapine, risperidone and haloperidol on the neuropathology (i.e. neuronal vacuolization) and the expression of Fos-like protein and c-fos mRNA in the retrosplenial cortex of female Sprague-Dawley rats induced by the NMDA receptor antagonist dizocilpine. Dizocilpine Maleate 296-307 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-169 10856442-3 2000 Furthermore, pretreatment (15 min) with clozapine or olanzapine, but not risperidone or haloperidol, significantly attenuated the expression of Fos-like protein in the retrosplenial cortex induced by dizocilpine (0.5 mg/kg, s.c.) in a dose-dependent manner. Clozapine 40-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-147 10856442-3 2000 Furthermore, pretreatment (15 min) with clozapine or olanzapine, but not risperidone or haloperidol, significantly attenuated the expression of Fos-like protein in the retrosplenial cortex induced by dizocilpine (0.5 mg/kg, s.c.) in a dose-dependent manner. Olanzapine 53-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-147 10856442-3 2000 Furthermore, pretreatment (15 min) with clozapine or olanzapine, but not risperidone or haloperidol, significantly attenuated the expression of Fos-like protein in the retrosplenial cortex induced by dizocilpine (0.5 mg/kg, s.c.) in a dose-dependent manner. Dizocilpine Maleate 200-211 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-147 10856442-4 2000 The marked expression of c-fos mRNA in the rat retrosplenial cortex induced by the administration of dizocilpine (0.5 mg/kg, s.c.) was significantly attenuated by pretreatment (15 min) with clozapine (10 mg/kg) or olanzapine (10 mg/kg), but not risperidone (10 mg/kg) or haloperidol (10 mg/kg). Dizocilpine Maleate 101-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 10856442-4 2000 The marked expression of c-fos mRNA in the rat retrosplenial cortex induced by the administration of dizocilpine (0.5 mg/kg, s.c.) was significantly attenuated by pretreatment (15 min) with clozapine (10 mg/kg) or olanzapine (10 mg/kg), but not risperidone (10 mg/kg) or haloperidol (10 mg/kg). Clozapine 190-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 10856442-4 2000 The marked expression of c-fos mRNA in the rat retrosplenial cortex induced by the administration of dizocilpine (0.5 mg/kg, s.c.) was significantly attenuated by pretreatment (15 min) with clozapine (10 mg/kg) or olanzapine (10 mg/kg), but not risperidone (10 mg/kg) or haloperidol (10 mg/kg). Olanzapine 214-224 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 10856442-4 2000 The marked expression of c-fos mRNA in the rat retrosplenial cortex induced by the administration of dizocilpine (0.5 mg/kg, s.c.) was significantly attenuated by pretreatment (15 min) with clozapine (10 mg/kg) or olanzapine (10 mg/kg), but not risperidone (10 mg/kg) or haloperidol (10 mg/kg). Risperidone 245-256 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 10856442-4 2000 The marked expression of c-fos mRNA in the rat retrosplenial cortex induced by the administration of dizocilpine (0.5 mg/kg, s.c.) was significantly attenuated by pretreatment (15 min) with clozapine (10 mg/kg) or olanzapine (10 mg/kg), but not risperidone (10 mg/kg) or haloperidol (10 mg/kg). Haloperidol 271-282 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 10856442-5 2000 The present results suggest that pharmacologically relevant doses of clozapine or olanzapine, but not risperidone or haloperidol, block the neuropathological changes and the expression of Fos-like protein and c-fos mRNA in the rat retrosplenial cortex elicited by the administration of dizocilpine. Clozapine 69-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 188-191 10856442-5 2000 The present results suggest that pharmacologically relevant doses of clozapine or olanzapine, but not risperidone or haloperidol, block the neuropathological changes and the expression of Fos-like protein and c-fos mRNA in the rat retrosplenial cortex elicited by the administration of dizocilpine. Clozapine 69-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 209-214 10856442-5 2000 The present results suggest that pharmacologically relevant doses of clozapine or olanzapine, but not risperidone or haloperidol, block the neuropathological changes and the expression of Fos-like protein and c-fos mRNA in the rat retrosplenial cortex elicited by the administration of dizocilpine. Olanzapine 82-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 188-191 10856442-5 2000 The present results suggest that pharmacologically relevant doses of clozapine or olanzapine, but not risperidone or haloperidol, block the neuropathological changes and the expression of Fos-like protein and c-fos mRNA in the rat retrosplenial cortex elicited by the administration of dizocilpine. Olanzapine 82-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 209-214 10856442-5 2000 The present results suggest that pharmacologically relevant doses of clozapine or olanzapine, but not risperidone or haloperidol, block the neuropathological changes and the expression of Fos-like protein and c-fos mRNA in the rat retrosplenial cortex elicited by the administration of dizocilpine. Dizocilpine Maleate 286-297 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 188-191 10825490-3 2000 Water induced little c-Fos-like immunoreactivity (c-FLI), but both intraoral and intragastric infusions of sucrose, but not non-caloric saccharin, induced strong c-FLI in the AP, caudal NTS and the external lateral subnucleus of the rostral PBN, suggesting that these areas receive general visceral inputs. Sucrose 107-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 10825502-3 2000 Fos induced by exposure of rats to fox urine or a neutral odor, mineral oil, was markedly enhanced during the subjective night compared to subjective day in the main olfactory bulb, primary olfactory cortex, and other structures related to olfaction. Mineral Oil 64-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 10888068-0 2000 Ethanol-induced c-fos expression in catecholamine- and neuropeptide Y-producing neurons in rat brainstem. Ethanol 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 10888068-0 2000 Ethanol-induced c-fos expression in catecholamine- and neuropeptide Y-producing neurons in rat brainstem. Catecholamines 36-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 10888068-1 2000 BACKGROUND: Previous studies have used c-Fos-like immunoreactivity (cFLI) to examine the neuroanatomical location of cells that are activated in response to ethanol administration. Ethanol 157-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 10888068-12 2000 CONCLUSIONS: Neurons in the rat brainstem that show ethanol-induced c-Fos expression produce catecholamines and NPY. Ethanol 52-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 10888068-12 2000 CONCLUSIONS: Neurons in the rat brainstem that show ethanol-induced c-Fos expression produce catecholamines and NPY. Catecholamines 93-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 10911868-0 2000 Molecular effects of the psychotropic NMDA receptor antagonist MK-801 in the rat entorhinal cortex: increases in AP-1 DNA binding activity and expression of Fos and Jun family members. Dizocilpine Maleate 63-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-160 10911868-4 2000 We also observed increased expression of mRNAs for Fos and Jun transcription factor family members c-Fos, FosB, Fra-2, and JunB, as well as Fos family proteins in the entorhinal cortex after MK-801 administration. Dizocilpine Maleate 191-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 10911868-4 2000 We also observed increased expression of mRNAs for Fos and Jun transcription factor family members c-Fos, FosB, Fra-2, and JunB, as well as Fos family proteins in the entorhinal cortex after MK-801 administration. Dizocilpine Maleate 191-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 10911868-4 2000 We also observed increased expression of mRNAs for Fos and Jun transcription factor family members c-Fos, FosB, Fra-2, and JunB, as well as Fos family proteins in the entorhinal cortex after MK-801 administration. Dizocilpine Maleate 191-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-104 10789833-3 2000 In addition, this work examined the effects of spinal hypothermia on FOS expression induced either by ischemia or by potassium-evoked depolarization (intrathecal KCl). Potassium 117-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 10789833-3 2000 In addition, this work examined the effects of spinal hypothermia on FOS expression induced either by ischemia or by potassium-evoked depolarization (intrathecal KCl). Potassium Chloride 162-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 10789833-11 2000 Intrathecal KCl-induced FOS expression in spinal neurons in the dorsal horn and in the intermediate zone. Potassium Chloride 12-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 10866986-7 2000 In a separate experiment using rats, delivery of carbonated water to the tongue significantly increased the number of cells expressing c-fos-like immunoreactivity in the dorsomedial trigeminal nucleus caudalis (versus saline controls); this was significantly reduced by pretreatment with acetazolamide. Water 60-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 10866986-7 2000 In a separate experiment using rats, delivery of carbonated water to the tongue significantly increased the number of cells expressing c-fos-like immunoreactivity in the dorsomedial trigeminal nucleus caudalis (versus saline controls); this was significantly reduced by pretreatment with acetazolamide. Acetazolamide 288-301 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 10913983-11 2000 Clozapine increases Fos protein immunoreactivity in the PFC with no or minimal effects in the DLSt. Clozapine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 10913983-12 2000 In contrast, haloperidol increases Fos protein immunoreactivity in the DLSt with minor effect in the PFC. Haloperidol 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 10913983-13 2000 Other atypical neuroleptics (risperidone, sertindole and NNC 22-0031) induced a Fos protein expression pattern different from haloperidol: The atypical compounds exhibit a larger ratio between Fos protein expression in PFC and DLSt than measured for haloperidol. Risperidone 29-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 10913983-13 2000 Other atypical neuroleptics (risperidone, sertindole and NNC 22-0031) induced a Fos protein expression pattern different from haloperidol: The atypical compounds exhibit a larger ratio between Fos protein expression in PFC and DLSt than measured for haloperidol. Risperidone 29-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-196 10913983-13 2000 Other atypical neuroleptics (risperidone, sertindole and NNC 22-0031) induced a Fos protein expression pattern different from haloperidol: The atypical compounds exhibit a larger ratio between Fos protein expression in PFC and DLSt than measured for haloperidol. sertindole 42-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 10913983-13 2000 Other atypical neuroleptics (risperidone, sertindole and NNC 22-0031) induced a Fos protein expression pattern different from haloperidol: The atypical compounds exhibit a larger ratio between Fos protein expression in PFC and DLSt than measured for haloperidol. sertindole 42-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-196 10913983-13 2000 Other atypical neuroleptics (risperidone, sertindole and NNC 22-0031) induced a Fos protein expression pattern different from haloperidol: The atypical compounds exhibit a larger ratio between Fos protein expression in PFC and DLSt than measured for haloperidol. O-[2-(1,3-Dioxo-1,3-Dihydro-2h-Isoindol-2-Yl)ethyl] (4-Chlorophenyl)thiocarbamate 57-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 10913983-13 2000 Other atypical neuroleptics (risperidone, sertindole and NNC 22-0031) induced a Fos protein expression pattern different from haloperidol: The atypical compounds exhibit a larger ratio between Fos protein expression in PFC and DLSt than measured for haloperidol. O-[2-(1,3-Dioxo-1,3-Dihydro-2h-Isoindol-2-Yl)ethyl] (4-Chlorophenyl)thiocarbamate 57-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-196 10913983-13 2000 Other atypical neuroleptics (risperidone, sertindole and NNC 22-0031) induced a Fos protein expression pattern different from haloperidol: The atypical compounds exhibit a larger ratio between Fos protein expression in PFC and DLSt than measured for haloperidol. Haloperidol 250-261 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-196 10886349-1 2000 This study examined whether conditioned hyperactivity measured in a cocaine-paired environment was associated with increased expression of Fos-related antigens (FRA) within the nucleus accumbens (NAc) and associated forebrain regions of rats. Cocaine 68-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 10799240-14 2000 The number of fos-protein positive neurons in the L6 spinal cord segment in the neuromodulation group (93.2 +/- 13.3 cells/section) decreased significantly when compared with the sham with acetic acid group (160.6 +/- 25.0 cells/section; p = 0.02). Acetic Acid 189-200 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 11956571-0 2000 [Increased expression of formalin-induced Fos and NADPH-d positive neurons in the spinal cord of morphine-tolerant rats]. Formaldehyde 25-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 11956571-0 2000 [Increased expression of formalin-induced Fos and NADPH-d positive neurons in the spinal cord of morphine-tolerant rats]. Morphine 97-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 11956571-1 2000 Fos immunocytochemistry, NADPH-d histochemistry and Fos/NADPH-d double-labeling method were used to study the changes in formalin-induced Fos, NADPH-d positive and Fos/NADPH-d double-labeled neurons in the spinal cord of morphine-tolerant rats. Formaldehyde 121-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 11956571-1 2000 Fos immunocytochemistry, NADPH-d histochemistry and Fos/NADPH-d double-labeling method were used to study the changes in formalin-induced Fos, NADPH-d positive and Fos/NADPH-d double-labeled neurons in the spinal cord of morphine-tolerant rats. Formaldehyde 121-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 11956571-1 2000 Fos immunocytochemistry, NADPH-d histochemistry and Fos/NADPH-d double-labeling method were used to study the changes in formalin-induced Fos, NADPH-d positive and Fos/NADPH-d double-labeled neurons in the spinal cord of morphine-tolerant rats. Formaldehyde 121-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 11956571-2 2000 Formalin-induced Fos-like immunoreactivity (Fos-LI) was located in the superficial laminae and neck of ipsilateral spinal cord. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 11956571-2 2000 Formalin-induced Fos-like immunoreactivity (Fos-LI) was located in the superficial laminae and neck of ipsilateral spinal cord. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 11956571-3 2000 Acute administration of morphine decreased the expression of Fos-LI in non-tolerant rats, while the expression of Fos-LI was significantly increased in morphine-tolerant rats. Morphine 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 11956571-3 2000 Acute administration of morphine decreased the expression of Fos-LI in non-tolerant rats, while the expression of Fos-LI was significantly increased in morphine-tolerant rats. Morphine 152-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 11956571-7 2000 A few formalin-induced Fos/NADPH-d double-labeled neurons were detected in the superficial laminae of spinal dorsal horn of non-tolerant rats. Formaldehyde 6-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 10819899-0 2000 Substantia nigra glutamate antagonists produce contralateral turning and basal ganglia Fos expression: interactions with D1 and D2 dopamine receptor agonists. Glutamic Acid 17-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 10819899-4 2000 The NMDA antagonist AP5 (1 microg), or the AMPA/kainate antagonist DNQX (0.015-1.5 microg), injected into the SNr (0.5 microl) elicited contralateral turning as well as both striatal and pallidal Fos expression. FG 9041 67-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 196-199 10819899-5 2000 Moreover, intranigral DNQX elicited more turning and greater numbers of Fos-positive striatal neurons in 6-OHDA-lesioned animals than in unlesioned controls, suggesting that the 6-OHDA injection induces functional changes in nigral glutamate transmission. FG 9041 22-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 10819899-5 2000 Moreover, intranigral DNQX elicited more turning and greater numbers of Fos-positive striatal neurons in 6-OHDA-lesioned animals than in unlesioned controls, suggesting that the 6-OHDA injection induces functional changes in nigral glutamate transmission. Oxidopamine 105-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 10819899-5 2000 Moreover, intranigral DNQX elicited more turning and greater numbers of Fos-positive striatal neurons in 6-OHDA-lesioned animals than in unlesioned controls, suggesting that the 6-OHDA injection induces functional changes in nigral glutamate transmission. Oxidopamine 178-184 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 10819899-7 2000 increased striatal Fos expression due to intranigral DNQX. FG 9041 53-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 10819899-11 2000 6-OHDA-lesioned rats given 5 microg of intrastriatal quinpirole exhibited both turning and pallidal Fos that was significantly increased by intranigral AP5. Oxidopamine 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-103 10819899-11 2000 6-OHDA-lesioned rats given 5 microg of intrastriatal quinpirole exhibited both turning and pallidal Fos that was significantly increased by intranigral AP5. Quinpirole 53-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-103 11956571-8 2000 In morphine-tolerant rats, on the other hand, formalin-induced Fos/NADPH-d double-labeled neurons were increased and distributed in the whole laminae of the ipsilateral spinal cord and the contralateral superficial spinal cord. NADP 67-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 11956571-9 2000 It is suggested that NO is involved in the increase of formalin-induced Fos-LI in the spinal cord of morphine-tolerant rats and may play an important role in the development of morphine tolerance. Formaldehyde 55-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 11956571-9 2000 It is suggested that NO is involved in the increase of formalin-induced Fos-LI in the spinal cord of morphine-tolerant rats and may play an important role in the development of morphine tolerance. Morphine 101-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 10891591-5 2000 Conversely, the inducible 62-65-kDa c-Fos is present in nuclear fractions from metrazole-treated animals (positive control), but not in Syn or SPM fractions. Pentylenetetrazole 79-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 10808081-3 2000 Here, inactivation of G(i/o)-proteins by pre-treatment of morphine-dependent rats with pertussis toxin injected into the left supraoptic nucleus reduced withdrawal-induced Fos protein expression within the injected nucleus by 41+/-10% compared to the contralateral nucleus, indicating that functional G(i/o)-proteins are essential for the development and/or expression of morphine dependence by oxytocin cells in the supraoptic nucleus. Morphine 58-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-175 10808081-5 2000 Finally, pertussis toxin reduced acute morphine inhibition of systemic hypertonic saline-induced Fos protein expression in the supraoptic nucleus, confirming that pertussis toxin effectively inactivates G(i/o)-proteins in the supraoptic nucleus. Morphine 39-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 10808081-5 2000 Finally, pertussis toxin reduced acute morphine inhibition of systemic hypertonic saline-induced Fos protein expression in the supraoptic nucleus, confirming that pertussis toxin effectively inactivates G(i/o)-proteins in the supraoptic nucleus. Sodium Chloride 82-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 11956571-8 2000 In morphine-tolerant rats, on the other hand, formalin-induced Fos/NADPH-d double-labeled neurons were increased and distributed in the whole laminae of the ipsilateral spinal cord and the contralateral superficial spinal cord. Morphine 3-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 11956571-8 2000 In morphine-tolerant rats, on the other hand, formalin-induced Fos/NADPH-d double-labeled neurons were increased and distributed in the whole laminae of the ipsilateral spinal cord and the contralateral superficial spinal cord. Formaldehyde 46-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 10781476-8 2000 The expression of c-fos mRNA was transiently increased in the brain, and more strikingly and for longer times, in the kidney with all three anesthetics; the expression of c-fos mRNA was decreased in the heart with isoflurane and pentobarbital and increased in the liver with isoflurane and propofol. Isoflurane 214-224 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-176 10781468-0 2000 The effect of diclofenac on the expression of spinal cord c-fos-like immunoreactivity after ischemia-reperfusion-induced acute hyperalgesia in the rat tail. Diclofenac 14-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 10781468-5 2000 After diclofenac pretreatment (subcutaneous 40 mg/Kg, 30 min before insult) the number of c-fos-LI-labeled neurons at 90 min was increased to 424% in laminae I-II, 150% in laminae III-IV, 142% in laminae V-X, and 183% in total (all P < 0.01). Diclofenac 6-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 10781468-6 2000 Thus diclofenac pretreatment partially prevented the insult-induced increase in total and regional neuronal c-fos-LI. Diclofenac 5-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 10781476-8 2000 The expression of c-fos mRNA was transiently increased in the brain, and more strikingly and for longer times, in the kidney with all three anesthetics; the expression of c-fos mRNA was decreased in the heart with isoflurane and pentobarbital and increased in the liver with isoflurane and propofol. Isoflurane 214-224 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 10831251-10 2000 Intragastric hypertonic saline infusion increases portal venous, but not systemic plasma, osmolality and increases Fos-like immunoreactivity in the AP, nucleus tractus solitarius and the supraoptic, paraventricular and lateral parabrachial nuclei. Sodium Chloride 24-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-118 10781476-8 2000 The expression of c-fos mRNA was transiently increased in the brain, and more strikingly and for longer times, in the kidney with all three anesthetics; the expression of c-fos mRNA was decreased in the heart with isoflurane and pentobarbital and increased in the liver with isoflurane and propofol. Pentobarbital 229-242 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 10781476-8 2000 The expression of c-fos mRNA was transiently increased in the brain, and more strikingly and for longer times, in the kidney with all three anesthetics; the expression of c-fos mRNA was decreased in the heart with isoflurane and pentobarbital and increased in the liver with isoflurane and propofol. Pentobarbital 229-242 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-176 10781476-8 2000 The expression of c-fos mRNA was transiently increased in the brain, and more strikingly and for longer times, in the kidney with all three anesthetics; the expression of c-fos mRNA was decreased in the heart with isoflurane and pentobarbital and increased in the liver with isoflurane and propofol. Isoflurane 275-285 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 10781476-8 2000 The expression of c-fos mRNA was transiently increased in the brain, and more strikingly and for longer times, in the kidney with all three anesthetics; the expression of c-fos mRNA was decreased in the heart with isoflurane and pentobarbital and increased in the liver with isoflurane and propofol. Isoflurane 275-285 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-176 10781476-8 2000 The expression of c-fos mRNA was transiently increased in the brain, and more strikingly and for longer times, in the kidney with all three anesthetics; the expression of c-fos mRNA was decreased in the heart with isoflurane and pentobarbital and increased in the liver with isoflurane and propofol. Propofol 290-298 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 10781476-8 2000 The expression of c-fos mRNA was transiently increased in the brain, and more strikingly and for longer times, in the kidney with all three anesthetics; the expression of c-fos mRNA was decreased in the heart with isoflurane and pentobarbital and increased in the liver with isoflurane and propofol. Propofol 290-298 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-176 10797155-4 2000 BAPTA, a Ca(2+) chelator, inhibited c-fos mRNA and promoter activation by hypoxia. 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 10804015-8 2000 In contrast to PTH, prostaglandin E2 (PGE2) had no effect on OPG mRNA expression in vivo in the metaphyseal bone cells, under conditions in which PGE2 does promote expression of the c-fos gene. Dinoprostone 146-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 182-187 10972462-1 2000 RATIONALE: We have previously shown that environmental novelty enhances the behavioral activating effects of amphetamine and amphetamine-induced expression of the immediate early gene c-fos in the striatal complex, particularly in the most caudal portion of the caudate. Amphetamine 109-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 184-189 10972462-1 2000 RATIONALE: We have previously shown that environmental novelty enhances the behavioral activating effects of amphetamine and amphetamine-induced expression of the immediate early gene c-fos in the striatal complex, particularly in the most caudal portion of the caudate. Amphetamine 125-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 184-189 10800959-8 2000 Site-specific interaction between the ligand and its receptor was further supported by the induction of c-fos-immunoreactive nuclei within EP4-expressing neurons in response to intracerebroventricular PGE2 infusion. Dinoprostone 201-205 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 10792606-11 2000 Immunoprecipitation of p300-C showed an interaction with the transcription factor c-Fos, which was enhanced by H2O2 treatment. Hydrogen Peroxide 111-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 10731628-0 2000 Carbamazepine suppresses methamphetamine-induced Fos expression in a regionally specific manner in the rat brain. Carbamazepine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 10731628-0 2000 Carbamazepine suppresses methamphetamine-induced Fos expression in a regionally specific manner in the rat brain. Methamphetamine 25-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 10731628-4 2000 To elucidate the neurobiological substrates responsible for the antimanic effects of carbamazepine, this study investigated the effects of chronic carbamazepine administration on regional Fos protein expression induced by a single dose of methamphetamine (2mg/kg). Carbamazepine 147-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 188-191 10731628-4 2000 To elucidate the neurobiological substrates responsible for the antimanic effects of carbamazepine, this study investigated the effects of chronic carbamazepine administration on regional Fos protein expression induced by a single dose of methamphetamine (2mg/kg). Methamphetamine 239-254 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 188-191 10731628-5 2000 Chronic treatment with CBZ (0.25% in food for 7 days, followed by 0.5% for 7 days; final mean serum carbamazepine concentration: 4.09 +/- 0.34 microg/ml) significantly attenuated the number of Fos-like immunoreactivity-positive nuclei induced by methamphetamine administration in the core of the nucleus accumbens and the caudate/putamen. Carbamazepine 23-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-196 10731628-5 2000 Chronic treatment with CBZ (0.25% in food for 7 days, followed by 0.5% for 7 days; final mean serum carbamazepine concentration: 4.09 +/- 0.34 microg/ml) significantly attenuated the number of Fos-like immunoreactivity-positive nuclei induced by methamphetamine administration in the core of the nucleus accumbens and the caudate/putamen. Carbamazepine 100-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-196 10797155-9 2000 KN-93, a CaMK inhibitor, blocked CaMKII activation and c-fos promoter stimulation by hypoxia. KN 93 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 10797155-13 2000 These results demonstrate that c-fos activation by hypoxia involves CaMK activation and CREB phosphorylation at Ser-133 and requires Ca/CRE and SRE. Serine 112-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 10837847-0 2000 Effect of acute nicotine on Fos protein expression in rat brain during chronic nicotine and its withdrawal. Nicotine 16-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 10837847-0 2000 Effect of acute nicotine on Fos protein expression in rat brain during chronic nicotine and its withdrawal. Nicotine 79-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 10837847-3 2000 In control rats, acute nicotine increased Fos IS significantly in all three brain areas studied. Nicotine 23-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 10837847-5 2000 After 72-h withdrawal nicotine-induced elevation of Fos IS was similar to that of control rats in all three areas. Nicotine 22-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 10751579-0 2000 Acute ethanol induction of c-Fos immunoreactivity in pre-pro-enkephalin expressing neurons of the central nucleus of the amygdala. Ethanol 6-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 10859491-15 2000 High concentrations of GnRH (1 microM) induced the appearance of a higher percent of LH-containing cells having c-fos than did 10 nM GnRH (p < 0.01), whereas a lower percent of LH-containing cells with c-fos were observed when the oxytocin concentration was raised from 10 nM to 1 microM (p < 0.02). Luteinizing Hormone 85-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 10767061-4 2000 In saline-treated rats, footshock resulted in an increase in Fos-li in the prelimbic and infralimbic cortices and tyrosine hydroxylase-labeled cells in the ventral tegmental area. Sodium Chloride 3-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 10767061-5 2000 Treatment with lorazepam or bretazenil prevented the stress-induced activation in Fos-li nuclei in all regions of the medial prefrontal cortex and in dopaminergic neurons in the ventral tegmental area. Lorazepam 15-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 10767061-5 2000 Treatment with lorazepam or bretazenil prevented the stress-induced activation in Fos-li nuclei in all regions of the medial prefrontal cortex and in dopaminergic neurons in the ventral tegmental area. bretazenil 28-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 10767061-8 2000 Additionally, treatment with R(+)HA-966 completely blocked, while guanfacine attenuated, the stress-induced increase in the number of Fos-li, TH-li cells in the ventral tegmental area. Guanfacine 66-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-137 10773195-5 2000 The accumbens site mediating morphine-induced increases in food "wanting" and "liking" was identified using a novel method based on local expression of Fos induced directly by drug microinjections. Morphine 29-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-155 10773195-6 2000 The plume-shaped region of drug-induced increase in Fos immunoreactivity immediately surrounding a morphine microinjection site (Fos plume) was objectively mapped. Morphine 99-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 10773195-6 2000 The plume-shaped region of drug-induced increase in Fos immunoreactivity immediately surrounding a morphine microinjection site (Fos plume) was objectively mapped. Morphine 99-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-132 10773195-7 2000 A point-sampling procedure was used to measure the shape and size of "positive" plumes of Fos expression triggered by microinjections of morphine at locations that caused increases in eating behavior. Morphine 137-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 10773422-0 2000 Inhibitory effect of intrathecally administered nociceptin on the expression of Fos-like immunoreactivity in the rat formalin test. Formaldehyde 117-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 10773422-2 2000 The authors examined the effect of 17 nmol of nociceptin on the expression of Fos-like immunoreactivity (Fos-LI) in the spinal dorsal horn induced by paw formalin injection and compared the effect of 17 nmol of nociceptin with that of equiantinociceptive dose (3 nmol) of morphine. Formaldehyde 154-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 10773422-2 2000 The authors examined the effect of 17 nmol of nociceptin on the expression of Fos-like immunoreactivity (Fos-LI) in the spinal dorsal horn induced by paw formalin injection and compared the effect of 17 nmol of nociceptin with that of equiantinociceptive dose (3 nmol) of morphine. Formaldehyde 154-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-108 10773422-4 2000 Both 17 nmol of nociceptin and 3 nmol of morphine suppressed the expression of Fos-LI in laminae I-II, but not in laminae III-V, to the same extent. Morphine 41-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 10751432-10 2000 Finally, BDNF caused phosphorylation of mitogen-activated protein kinase (MAPK), and because the treatment with the MAPK inhibitor U0126 completely abolished CREB activation and c-fos upregulation, it is likely that both processes depend mainly on the MAP kinase pathway. U 0126 131-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 178-183 10754219-0 2000 Differential activation of Fos-like immunoreactivity in the arcuate nucleus and amygdala after intracerebroventricular injection of sodium nitroprusside and N omega nitro-L-arginine in conscious and urethane-anesthetized lactating rats. Nitroprusside 132-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 10754219-0 2000 Differential activation of Fos-like immunoreactivity in the arcuate nucleus and amygdala after intracerebroventricular injection of sodium nitroprusside and N omega nitro-L-arginine in conscious and urethane-anesthetized lactating rats. Nitroarginine 157-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 10754219-0 2000 Differential activation of Fos-like immunoreactivity in the arcuate nucleus and amygdala after intracerebroventricular injection of sodium nitroprusside and N omega nitro-L-arginine in conscious and urethane-anesthetized lactating rats. Urethane 199-207 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 10814831-7 2000 The c-fos mRNA induced by forskolin, but not by NGF, was also suppressed by Puralpha transfection. Colforsin 26-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 10814832-0 2000 Prior experience of morphine application alters the c-fos response to MDMA ("ecstasy") and cocaine in the rat striatum. Morphine 20-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 10814832-0 2000 Prior experience of morphine application alters the c-fos response to MDMA ("ecstasy") and cocaine in the rat striatum. N-Methyl-3,4-methylenedioxyamphetamine 70-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 10814832-0 2000 Prior experience of morphine application alters the c-fos response to MDMA ("ecstasy") and cocaine in the rat striatum. Cocaine 91-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 10814832-7 2000 MDMA (3, 4-methylenedioxymethamphetamine, 6 mg/kg) as a single test dose yielded a c-fos response in a wide range of brain areas. N-Methyl-3,4-methylenedioxyamphetamine 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 10814832-7 2000 MDMA (3, 4-methylenedioxymethamphetamine, 6 mg/kg) as a single test dose yielded a c-fos response in a wide range of brain areas. N-Methyl-3,4-methylenedioxyamphetamine 6-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 10814832-8 2000 In the caudate putamen, the expression pattern of c-fos was clearly altered if the rats had received repeated morphine application previously. Morphine 110-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 10814832-9 2000 In this case, the MDMA-induced c-fos expression was markedly confined to the centromedial, mesolimbic aspect of the striatum whereas it had a diffuse appearance in rats not exposed to the opiate earlier. N-Methyl-3,4-methylenedioxyamphetamine 18-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 10814832-10 2000 Cocaine application (50 mg/kg) elicited an intense c-fos expression in the medial striatum if the animals were morphine-pretreated; it was virtually absent in drug-naive rats after the same cocaine dose. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 10814832-10 2000 Cocaine application (50 mg/kg) elicited an intense c-fos expression in the medial striatum if the animals were morphine-pretreated; it was virtually absent in drug-naive rats after the same cocaine dose. Morphine 111-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 10760498-6 2000 In both experiments, the furosemide-treated rats had significantly more Fos-positive cell nuclei than vehicle-treated rats in the subfornical organ (SFO), organum vasculosum lamina terminalis (OVLT), supraoptic nuclei (SON), and magnocellular region of the paraventricular nuclei (PVN) - areas previously shown to be activated by hypovolemia or peripheral angiotensin. Furosemide 25-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 10760498-7 2000 In the short-survival experiment, the furosemide-treated rats had more Fos-positive cell nuclei in the nucleus of the solitary tract (NTS) and in the dorsal horn of the spinal cord at spinal levels T(11), T(12), and T(13). Furosemide 38-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 10760498-12 2000 In this experiment, furosemide administration increased the number of Fos-positive cells in the SFO, OVLT, SON and PVN, but not in the caudal thoracic spinal cord or NTS. Furosemide 20-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 10760499-11 2000 Rats receiving furosemide plus the low CAP dose showed more Fos-positive cells than control rats in the subfornical organ (SFO), organum vasculosum lamina terminalis (OVLT), supraoptic nucleus (SON), magnocellular region of the paraventricular nucleus, nucleus of the solitary tract (NTS), and caudal thoracic/rostral lumbar spinal cord dorsal horn. Furosemide 15-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 10766418-8 2000 Previously, we found that a serum response factor is activated by copper in the LEC rat liver, and suggested that it must mediate proto-oncogene c-fos induction. Copper 66-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-150 10766418-9 2000 The results obtained here suggest that accumulation of diacylglycerol plays an important role in development of hepatoma in LEC rats by mediating proto-oncogene c-fos induction. Diglycerides 55-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 10749792-1 2000 These studies examined Fos protein expression in spinal cord neurons synaptically activated by stimulation of bladder afferent pathways after cyclophosphamide (CYP)-induced bladder inflammation. Cyclophosphamide 142-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 10749792-2 2000 In urethan-anesthetized Wistar rats with cystitis, intravesical saline distension significantly (P </= 0.0005) increased the number of Fos-immunoreactive (IR) cells observed in the rostral lumbar (L1, 35 cells/section; L2, 27 cells/section) and caudal lumbosacral (L6, 120 cells/section; S1, 96 cells/section) spinal cord compared with control animals, but Fos protein expression in the L5 segment was not altered. Urethane 3-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-141 10749792-2 2000 In urethan-anesthetized Wistar rats with cystitis, intravesical saline distension significantly (P </= 0.0005) increased the number of Fos-immunoreactive (IR) cells observed in the rostral lumbar (L1, 35 cells/section; L2, 27 cells/section) and caudal lumbosacral (L6, 120 cells/section; S1, 96 cells/section) spinal cord compared with control animals, but Fos protein expression in the L5 segment was not altered. Sodium Chloride 64-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-141 10749792-2 2000 In urethan-anesthetized Wistar rats with cystitis, intravesical saline distension significantly (P </= 0.0005) increased the number of Fos-immunoreactive (IR) cells observed in the rostral lumbar (L1, 35 cells/section; L2, 27 cells/section) and caudal lumbosacral (L6, 120 cells/section; S1, 96 cells/section) spinal cord compared with control animals, but Fos protein expression in the L5 segment was not altered. Sodium Chloride 64-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 360-363 10749792-7 2000 Pretreatment with capsaicin significantly reduced the number of Fos-IR cells induced by bladder distension after cystitis. Capsaicin 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 10749792-8 2000 These data suggest that chronic cystitis can reveal a nociceptive Fos expression pattern in the spinal cord in response to a non-noxious bladder stimulus that is partially mediated by capasaicin-sensitive bladder afferents. capasaicin 184-194 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 10758334-0 2000 Behavioral and c-fos expression changes induced by nitric oxide donors microinjected into the dorsal periaqueductal gray. Nitric Oxide 51-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 10716675-0 2000 The 5-HT(3) receptor antagonist alosetron inhibits the colorectal distention induced depressor response and spinal c-fos expression in the anaesthetised rat. alosetron 32-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 10716675-6 2000 Rats anaesthetised with urethane and treated with alosetron or saline underwent a repeated CRD paradigm, after which the lumbosacral spinal cord was removed and processed for visualisation of Fos-LI. alosetron 50-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 192-195 10716675-10 2000 Pretreatment with alosetron (100 microg/kg) significantly reduced numbers of Fos-LI neurones to 479.8. alosetron 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-80 10864414-0 2000 Ciproxifan and cimetidine modulate c-fos expression in septal neurons, and acetylcholine release from hippocampus of freely moving rats. ciproxifan 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 10864414-0 2000 Ciproxifan and cimetidine modulate c-fos expression in septal neurons, and acetylcholine release from hippocampus of freely moving rats. Cimetidine 15-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 10737591-0 2000 Interaction between the serotoninergic and dopaminergic systems in d-fenfluramine-induced activation of c-fos and jun B genes in rat striatal neurons. Dexfenfluramine 67-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 10737591-2 2000 To determine the neuronal targets of d-fenfluramine in the striatum, we identified the phenotypes of striatal neurons in which d-fenfluramine induced Fos expression. Dexfenfluramine 37-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-153 10737591-2 2000 To determine the neuronal targets of d-fenfluramine in the striatum, we identified the phenotypes of striatal neurons in which d-fenfluramine induced Fos expression. Dexfenfluramine 127-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-153 10737591-3 2000 Our results demonstrated that d-fenfluramine evokes nuclear expression of Fos/Jun B proteins in the striatum, and that the Fos expression was dose-dependent and accompanied by transient induction of c-fos mRNA. Dexfenfluramine 30-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 10737591-3 2000 Our results demonstrated that d-fenfluramine evokes nuclear expression of Fos/Jun B proteins in the striatum, and that the Fos expression was dose-dependent and accompanied by transient induction of c-fos mRNA. Dexfenfluramine 30-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-126 10737591-3 2000 Our results demonstrated that d-fenfluramine evokes nuclear expression of Fos/Jun B proteins in the striatum, and that the Fos expression was dose-dependent and accompanied by transient induction of c-fos mRNA. Dexfenfluramine 30-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-204 10737591-4 2000 Fos expression was blocked by p-chloroamphetamine, a serotoninergic neurotoxin. p-Chloroamphetamine 30-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 10737591-5 2000 Pretreatment with SCH 23390, a D1-dopamine receptor antagonist, led to a marked decrease in Fos/Jun B expression in the caudoputamen, but not in the cortex, whereas pretreatment with methiothepin, a nonselective serotonin 5-HT1 receptor antagonist, blocked Fos expression completely in the cortex and only partially in the caudoputamen. SCH 23390 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 10737591-5 2000 Pretreatment with SCH 23390, a D1-dopamine receptor antagonist, led to a marked decrease in Fos/Jun B expression in the caudoputamen, but not in the cortex, whereas pretreatment with methiothepin, a nonselective serotonin 5-HT1 receptor antagonist, blocked Fos expression completely in the cortex and only partially in the caudoputamen. SCH 23390 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 257-260 10737591-8 2000 These results suggest that the mechanism by which d-fenfluramine induces c-fos and jun B expression in the rat caudoputamen depends at least in part on activation of the dopaminergic system by serotonin. Dexfenfluramine 50-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 10737591-8 2000 These results suggest that the mechanism by which d-fenfluramine induces c-fos and jun B expression in the rat caudoputamen depends at least in part on activation of the dopaminergic system by serotonin. Serotonin 193-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 10771495-0 2000 Fedotozine, a kappa-opioid agonist, prevents spinal and supra-spinal Fos expression induced by a noxious visceral stimulus in the rat. fedotozine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 10771495-9 2000 Capsaicin pretreatment blocked AA-induced Fos in all structures tested. Capsaicin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 10771495-10 2000 Fedotozine significantly decreased AA-induced abdominal cramps and Fos immunoreactivity in the spinal cord and PVN, this effect being reversed by nor-BNI pretreatment. fedotozine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 10771206-11 2000 Nicotine slightly increased both c-fos and c-jun mRNA level and GTS, which did not affect the basal c-fos and c-jun mRNA expression, further enhanced nicotine-induced c-fos and c-jun mRNA level at both VTA and NA regions. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 10771206-13 2000 GTS may exert an potentiative effect on both c-fos and c-jun mRNA expression at NA region through inhibiting the release of DA in NA. Glutathionesulfonic acid 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 10790876-0 2000 Serotonin depletion decreases light induced c-fos in the rat suprachiasmatic nucleus. Serotonin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 10790876-4 2000 Serotonin depletion, verified by immunohistochemistry, produced a significant decrease (42%) in the number of c-FOS positive cells in the ventrolateral portion of the SCN. Serotonin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 10751579-15 2000 Bloom, Acute ethanol induces c-fos immunoreactivity in GABAergic neurons of the central nucleus of the amygdala, Brain Res, 798 (1998) 333-336]. Ethanol 13-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 10751579-16 2000 In the present study, we report that ethanol-induced c-Fos immunoreactivity was mainly confined to neurons that express pro-enkephalin (ENK). Ethanol 37-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 11961588-0 2000 [Increased expression of c-fos in the brainstem nuclei involved in cardiova scular regulation by capsaicin]. Capsaicin 97-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 11961588-1 2000 Effects of intracarotid injected capsaicin on the expression of Fos proto-oncogene in the brainstem nuclei involved in cardiovascular regulation were examined in 16 anesthetized rats with sinoaortic denervation. Capsaicin 33-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 10700011-4 2000 At concentrations of 3 and 30 nM, respectively, (+/-)-7-OH-DPAT and PD 128,907 significantly increased the expression of c-fos mRNA. 7-hydroxy-2-N,N-dipropylaminotetralin 48-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-126 10700028-0 2000 3,4-Methylenedioxymetamphetamine (ecstasy) induces c-fos-like protein and mRNA in rat organotypic dorsal striatal slices. N-Methyl-3,4-methylenedioxyamphetamine 0-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 10700028-0 2000 3,4-Methylenedioxymetamphetamine (ecstasy) induces c-fos-like protein and mRNA in rat organotypic dorsal striatal slices. N-Methyl-3,4-methylenedioxyamphetamine 34-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 10700028-3 2000 In this study the effects of MDMA on the expression of c-fos mRNA by in situ hybridization as well as the c-fos-like protein by immunohistochemistry in isolated dorsal striatum was investigated. N-Methyl-3,4-methylenedioxyamphetamine 29-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 10700028-4 2000 It was shown that 100 microM MDMA induced c-fos mRNA expression 30 min after treatment. N-Methyl-3,4-methylenedioxyamphetamine 29-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 10700028-6 2000 The c-fos expression was inhibited by MK 801 and metoclopramide, indicating the involvement of dopaminergic D2 receptors and glutamatergic NMDA receptors. Dizocilpine Maleate 38-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 10700028-6 2000 The c-fos expression was inhibited by MK 801 and metoclopramide, indicating the involvement of dopaminergic D2 receptors and glutamatergic NMDA receptors. Metoclopramide 49-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 10700028-8 2000 We conclude that MDMA treatment leads to the induction of c-fos expression in isolated rat striatal slices. N-Methyl-3,4-methylenedioxyamphetamine 17-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 10700011-4 2000 At concentrations of 3 and 30 nM, respectively, (+/-)-7-OH-DPAT and PD 128,907 significantly increased the expression of c-fos mRNA. Palladium 68-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-126 10700011-5 2000 The action of (+/-)-7-OH-DPAT was expressed stereospecifically; its (+)-isomer (K(i) values, D(3)/D(2) = 1.6/56.7 elicited a 26% +/- 7.6% increase in c-fos expression whereas its (-)-isomer (K(i) values, D(3)/D(2) = 59/1,060 nM) was ineffective. 7-hydroxy-2-N,N-dipropylaminotetralin 14-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-155 10700011-8 2000 An examination of the concentration-response relationship revealed that (+/-)-7-OH-DPAT and PD 128,907 both produced bell-shaped dose-response curves for c-fos induction. 7-hydroxy-2-N,N-dipropylaminotetralin 72-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 10700011-8 2000 An examination of the concentration-response relationship revealed that (+/-)-7-OH-DPAT and PD 128,907 both produced bell-shaped dose-response curves for c-fos induction. Palladium 92-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 10700011-9 2000 The sequential activation of D(2) receptors-which inhibit striatal c-fos expression (Simpson and Morris [1995] Neuroscience 68:97-106)-by higher concentrations of (+/-)-7-OH-DPAT and PD 128,907 is presumably involved in the inflexion at higher doses. 7-hydroxy-2-N,N-dipropylaminotetralin 163-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 10776677-7 2000 These results show that (1) chronic alcohol consumption desensitizes or decreases the DAT of DA terminals in the ACC and that (2) EtOH causes cellular hyperexcitability of ACC dopaminergic neurons with increased Fos expression during alcohol tolerance. Ethanol 130-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 212-215 10686408-5 2000 In normal males and females, formalin increased c-Fos in the dorsal DG and in the male ventral subfields. Formaldehyde 29-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 10700605-5 2000 Conversely, Fos-positive GnRH neurons were rarely observed in d16, and some Fos-positive GnRH neurons were observed in the d30 group (p<0.05 vs. saline) and the mature group (p<0.01 vs. saline). Sodium Chloride 148-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 10663966-0 2000 Differential c-Fos and caspase expression following kainic acid excitotoxicity. Kainic Acid 52-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 10776677-0 2000 Changes in dopamine transporter and c-Fos expression in the nucleus accumbens of alcohol-tolerant rats. Alcohols 81-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 10776677-6 2000 Increased expression of the c-Fos-like protein was found in the ACC of alcohol-treated rats. Alcohols 71-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 10825686-6 2000 Expression of c-fos-like proteins was shown after the injection of vegetable oil and 5% mustard in vegetable oil (within 2 h) in the spinal cord structures, the maximum quantity of the labeled cells was found in the I and II sacral segments (in the superficial lamina of the gray matter of the dorsal horns in the ipsilateral side), the minimum in the IV and V lumbar segments (in the intermediate zone of the gray matter at the level of the central canal). Oils 77-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 10825686-6 2000 Expression of c-fos-like proteins was shown after the injection of vegetable oil and 5% mustard in vegetable oil (within 2 h) in the spinal cord structures, the maximum quantity of the labeled cells was found in the I and II sacral segments (in the superficial lamina of the gray matter of the dorsal horns in the ipsilateral side), the minimum in the IV and V lumbar segments (in the intermediate zone of the gray matter at the level of the central canal). Oils 109-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 10708732-8 2000 administration of M&B28,767 (1 pg/rat) attenuated the elevation of c-fos mRNA during naloxone-precipitated withdrawal in many brain regions, including the cerebral cortex, thalamus, hypothalamus and locus coeruleus. Adenosine Monophosphate 20-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 10708732-8 2000 administration of M&B28,767 (1 pg/rat) attenuated the elevation of c-fos mRNA during naloxone-precipitated withdrawal in many brain regions, including the cerebral cortex, thalamus, hypothalamus and locus coeruleus. Naloxone 89-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 10712293-0 2000 Sucrose consumption increases naloxone-induced c-Fos immunoreactivity in limbic forebrain. Sucrose 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 10712293-0 2000 Sucrose consumption increases naloxone-induced c-Fos immunoreactivity in limbic forebrain. Naloxone 30-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 10712293-6 2000 In the central nucleus of the amygdala, naloxone administration to those rats drinking water significantly increased c-Fos-IR, an effect that was significantly enhanced by sucrose consumption, suggesting an upregulation of endogenous opioid tone in this area. Naloxone 40-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 10712293-6 2000 In the central nucleus of the amygdala, naloxone administration to those rats drinking water significantly increased c-Fos-IR, an effect that was significantly enhanced by sucrose consumption, suggesting an upregulation of endogenous opioid tone in this area. Water 87-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 10712293-6 2000 In the central nucleus of the amygdala, naloxone administration to those rats drinking water significantly increased c-Fos-IR, an effect that was significantly enhanced by sucrose consumption, suggesting an upregulation of endogenous opioid tone in this area. Sucrose 172-179 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 10696082-3 2000 We also found that increases in sympathetic and cardiovascular activities by microinjection of L-glutamate into the rostral ventrolateral medulla are mediated by c-fos expression-related substance(s) following activation of the nitric oxide-cyclic GMP pathway. Glutamic Acid 95-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 10696082-3 2000 We also found that increases in sympathetic and cardiovascular activities by microinjection of L-glutamate into the rostral ventrolateral medulla are mediated by c-fos expression-related substance(s) following activation of the nitric oxide-cyclic GMP pathway. Nitric Oxide 228-240 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 10696082-3 2000 We also found that increases in sympathetic and cardiovascular activities by microinjection of L-glutamate into the rostral ventrolateral medulla are mediated by c-fos expression-related substance(s) following activation of the nitric oxide-cyclic GMP pathway. Cyclic GMP 241-251 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 10696082-8 2000 In addition, increases in arterial pressure, heart rate, and sympathetic nerve activity by pulsatile compression were significantly reduced 6 h after microinjection of antisense oligodeoxynucleotide to c-fos mRNA (2+/-2 mmHg, 2+/-1 b.p.m., 1.0+/-1.0%; P<0.05 vs sense oligodeoxynucleotide). Oligodeoxyribonucleotides 178-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 202-207 10696082-8 2000 In addition, increases in arterial pressure, heart rate, and sympathetic nerve activity by pulsatile compression were significantly reduced 6 h after microinjection of antisense oligodeoxynucleotide to c-fos mRNA (2+/-2 mmHg, 2+/-1 b.p.m., 1.0+/-1.0%; P<0.05 vs sense oligodeoxynucleotide). Oligodeoxyribonucleotides 271-291 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 202-207 10690748-0 2000 Flurothyl-induced seizures in rats activate Fos in brainstem catecholaminergic neurons. Flurothyl 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 10762327-5 2000 The decrease in dopamine observed in the shell had a postsynaptic impact, as shown by less induction of Fos-like proteins selectively in the shell in response to cocaine. Dopamine 16-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 10762327-5 2000 The decrease in dopamine observed in the shell had a postsynaptic impact, as shown by less induction of Fos-like proteins selectively in the shell in response to cocaine. Cocaine 162-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 10762327-6 2000 However, the induction of Fos-like proteins by the full D1 agonist SKF82958 (1.5 mg/kg, i.p.) SK and F 82958 67-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 10762327-7 2000 remained unchanged after adrenalectomy, suggesting that the changes in Fos expression after cocaine injection were likely to depend on changes in extracellular dopamine levels rather than on changes in postsynaptic sensitivity to dopamine. Cocaine 92-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 10762327-7 2000 remained unchanged after adrenalectomy, suggesting that the changes in Fos expression after cocaine injection were likely to depend on changes in extracellular dopamine levels rather than on changes in postsynaptic sensitivity to dopamine. Dopamine 160-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 10723097-0 2000 ATP-stimulated c-fos and zif268 mRNA expression is inhibited by chemical hypoxia in a rat brain-derived type 2 astrocyte cell line, RBA-2. Adenosine Triphosphate 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 10723097-1 2000 The stimulus-transcriptional coupling during ischemia/hypoxia was examined for ATP-stimulated expression of immediate early genes (IEGs; c-fos, zif268, c-myc and nur77) in a rat brain-derived type 2 astrocyte cell line, RBA-2. Adenosine Triphosphate 79-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 10723097-2 2000 Incubation of cells with 1 mM of extracellular ATP stimulated time-dependent expression of c-fos and zif268. Adenosine Triphosphate 47-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 10723097-3 2000 ATP induced the largest increases in zif268 mRNA and a lesser one in c-fos mRNA. Adenosine Triphosphate 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 10723097-5 2000 Brief exposure of cells to potassium cyanide to simulate chemical hypoxia induced 9-fold and 7-fold transient increases in c-fos and zif268 expression, respectively, but did not affect c-myc or nur77 expression. Potassium Cyanide 27-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 10723097-6 2000 When cyanide and ATP were added together, the expression of c-fos and zif268 expression was inhibited, and the effect was mimicked by simulating chemical hypoxia with sodium azide. Cyanides 5-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 10723097-6 2000 When cyanide and ATP were added together, the expression of c-fos and zif268 expression was inhibited, and the effect was mimicked by simulating chemical hypoxia with sodium azide. Adenosine Triphosphate 17-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 10723097-6 2000 When cyanide and ATP were added together, the expression of c-fos and zif268 expression was inhibited, and the effect was mimicked by simulating chemical hypoxia with sodium azide. Sodium Azide 167-179 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 10723097-9 2000 Nevertheless, metabolic inhibitor, iodoacetate, blocked the ATP-induced c-fos and partially inhibited zif268 expression, and deprivation of cells with glucose also inhibited the ATP-induced c-fos expression. Iodoacetates 35-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 10723097-9 2000 Nevertheless, metabolic inhibitor, iodoacetate, blocked the ATP-induced c-fos and partially inhibited zif268 expression, and deprivation of cells with glucose also inhibited the ATP-induced c-fos expression. Iodoacetates 35-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 190-195 10723097-9 2000 Nevertheless, metabolic inhibitor, iodoacetate, blocked the ATP-induced c-fos and partially inhibited zif268 expression, and deprivation of cells with glucose also inhibited the ATP-induced c-fos expression. Adenosine Triphosphate 60-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 10723097-9 2000 Nevertheless, metabolic inhibitor, iodoacetate, blocked the ATP-induced c-fos and partially inhibited zif268 expression, and deprivation of cells with glucose also inhibited the ATP-induced c-fos expression. Adenosine Triphosphate 60-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 190-195 10723097-9 2000 Nevertheless, metabolic inhibitor, iodoacetate, blocked the ATP-induced c-fos and partially inhibited zif268 expression, and deprivation of cells with glucose also inhibited the ATP-induced c-fos expression. Glucose 151-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 190-195 10723097-9 2000 Nevertheless, metabolic inhibitor, iodoacetate, blocked the ATP-induced c-fos and partially inhibited zif268 expression, and deprivation of cells with glucose also inhibited the ATP-induced c-fos expression. Adenosine Triphosphate 178-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 190-195 10723097-10 2000 Taken together, these results demonstrate that both extracellular ATP and chemical hypoxia induce c-fos and zif268 expression in RBA-2 type 2 astrocytes. Adenosine Triphosphate 66-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 10723097-11 2000 The chemical hypoxia inhibited ATP-stimulated c-fos and zif268 expression is not due to alterations in Ca(2+) and PLD signaling, and is at least partially related to metabolic disturbance in these cells. Adenosine Triphosphate 31-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 10693946-3 2000 Significant increases in the levels of c-fos, c-jun, and egr-1 but not NGFIB mRNA were observed in PC12 cells exposed to lead or phorbol 12-myristate 13-acetate. Tetradecanoylphorbol Acetate 129-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 10692604-0 2000 Morphine and NMDA receptor antagonism reduce c-fos expression in spinal trigeminal nucleus produced by acute injury to the TMJ region. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 10692604-12 2000 Combined subthreshold doses of morphine and MK-801 reduced c-fos expression in the dPa5, mid-Vc, and the Vc/C2 transition region, below that predicted from the effects of either drug alone. Morphine 31-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 10692604-12 2000 Combined subthreshold doses of morphine and MK-801 reduced c-fos expression in the dPa5, mid-Vc, and the Vc/C2 transition region, below that predicted from the effects of either drug alone. Dizocilpine Maleate 44-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 10700558-1 2000 Dual immunohistochemistry was employed to determine the effects of prolactin on expression of Fos and its related antigens (FRA) in tuberoinfundibular dopamine (TIDA) neurons located in the dorsomedial (DM) and ventrolateral (VL) subdivisions of the arcuate nucleus (ARC) in the male rat. Dopamine 151-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 10700558-1 2000 Dual immunohistochemistry was employed to determine the effects of prolactin on expression of Fos and its related antigens (FRA) in tuberoinfundibular dopamine (TIDA) neurons located in the dorsomedial (DM) and ventrolateral (VL) subdivisions of the arcuate nucleus (ARC) in the male rat. tida 161-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 10677633-1 2000 The present study investigated the effect of clonidine on the basal and inducible c-jun and c-fos mRNA expression in the nucleus tractus solitarius (middle, mNTS, and rostral, rNTS) and the rostral ventrolateral medulla (caudal, cRVLM, and rostral, rRVLM). Clonidine 45-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 10677633-5 2000 Clonidine attenuated the increases in c-fos in the mNTS and cRVLM and c-jun gene expression in the mNTS and rRVLM caused by NP-evoked hypotension and also reduced the basal expression of c-jun mRNA in the mNTS and rRVLM. Clonidine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 10677633-6 2000 These findings establish a causal link between clonidine inhibition of c-fos expression in brainstem and its hypotensive action, and provide the first evidence that clonidine attenuates the expression of the closely linked c-jun gene in neurons implicated in centrally mediated hypotension. Clonidine 47-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 10650129-3 2000 injection of CRF and urocortin in conscious rats by monitoring Fos expression 60 min later. Urocortins 21-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 10650136-0 2000 Nitric oxide synthase and glutamate receptor immunoreactivity in the rat spinal trigeminal neurons expressing Fos protein after formalin injection. Formaldehyde 128-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 10650136-2 2000 In this study, c-fos expression indicated by Fos immunohistochemistry in the caudal spinal trigeminal nucleus induced by subcutaneous injection of formalin into the lateral face of the rat was used as a marker for nociceptive neurons. Formaldehyde 147-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 10650136-2 2000 In this study, c-fos expression indicated by Fos immunohistochemistry in the caudal spinal trigeminal nucleus induced by subcutaneous injection of formalin into the lateral face of the rat was used as a marker for nociceptive neurons. Formaldehyde 147-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 10650136-4 2000 After formalin injection, many Fos-positive nuclei appeared in the superficial laminae of the ipsilateral trigeminal nucleus. Formaldehyde 6-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 10650136-7 2000 Lastly, some of the Fos-positive neurons were labelled by tetramethylrhodamine-dextran injected into the trigeminothalamic tract or the thalamic region. Fluoro-Ruby 58-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 10650136-8 2000 The results suggested that activation of N-methyl-D-aspartate receptor 1 and glutamate receptor 2/3 upon glutamate release in response to noxious stimulation to the orofacial region might mediate c-fos expression in neurons involved in nociception. Glutamic Acid 77-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 196-201 10650136-9 2000 The expression of Fos in the neurons could also be mediated by nitric oxide produced from the same, as well as neighbouring neurons, when nociceptive stimulation persisted. Nitric Oxide 63-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 10674475-6 2000 Severe hypoxia, where cytochrome oxidase was reduced, caused a significant induction of c-fos mRNA At this stage, the oxygen concentration in cerebral tissue fell to < 10(-7) M. These data suggest that the decline in oxidative phosphorylation might be a trigger for the induction of c-fos mRNA. Oxygen 118-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 10666140-0 2000 Dexfenfluramine and norfenfluramine: comparison of mechanism of action in feeding and brain Fos-ir studies. Dexfenfluramine 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 10666140-7 2000 Compared with controls, 5, 7-DHT-lesion rats showed greatly increased dF- and dNF-induced Fos-like immunoreactivity (ir) in the paraventricular (PVN) and supraoptic (SON) hypothalamic nuclei, and in the median preoptic area (MnPO), but were similar to controls in most other areas. 5,7-Dihydroxytryptamine 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 10666140-7 2000 Compared with controls, 5, 7-DHT-lesion rats showed greatly increased dF- and dNF-induced Fos-like immunoreactivity (ir) in the paraventricular (PVN) and supraoptic (SON) hypothalamic nuclei, and in the median preoptic area (MnPO), but were similar to controls in most other areas. Norfenfluramine 78-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 10666140-8 2000 PCPA pretreatment increased dF- and dNF-induced Fos-ir in the PVN, SON, and MnPO. Fenclonine 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 10666140-9 2000 In controls, equianorectic doses of dF and dNF induced Fos-ir in similar brain regions, but dNF produced relatively larger effects than dF in SON, PVN, and MnPO. Norfenfluramine 43-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 10719265-4 2000 In various parts of the forebrain and brainstem, BDA-labeled fibers originating from the cortex were observed in close apposition to Fos-like immunoreactive cells (FLI) activated by stimulation. biotinylated dextran amine 49-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-136 10650960-1 2000 During the preovulatory and estradiol-progesterone-induced GnRH-LH surge, a subpopulation of GnRH neurons transiently expresses the transcription factor c-fos, which is a useful marker of cell activation. Estradiol 28-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-158 10650960-1 2000 During the preovulatory and estradiol-progesterone-induced GnRH-LH surge, a subpopulation of GnRH neurons transiently expresses the transcription factor c-fos, which is a useful marker of cell activation. Progesterone 38-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-158 10650960-5 2000 The results show that the KA2-containing GnRH neurons also contain GluR5 receptor subunit mRNA and protein, and that these GnRH neurons are c-Fos positive during the steroid-induced LH surge. Steroids 166-173 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 10712794-4 2000 Non-toxic concentrations of anisomycin were found to stimulate these enzyme activities as well as the expression of the early response genes c-jun, c-fos and zif268, and to inhibit NGF-induced neurite formation. Anisomycin 28-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 10640314-0 2000 Intrathecally administered gabapentin inhibits formalin-evoked nociception and the expression of Fos-like immunoreactivity in the spinal cord of the rat. Gabapentin 27-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 10640314-5 2000 Unlike its inhibition of formalin-evoked nociceptive behaviors, the effect of gabapentin on the expression of Fos-like immunoreactivity in the spinal cord was highly dependent on the concentration of formalin. Gabapentin 78-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 10640314-5 2000 Unlike its inhibition of formalin-evoked nociceptive behaviors, the effect of gabapentin on the expression of Fos-like immunoreactivity in the spinal cord was highly dependent on the concentration of formalin. Formaldehyde 200-208 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 10648731-0 2000 Local morphine withdrawal increases c-fos gene, Fos protein, and oxytocin gene expression in hypothalamic magnocellular neurosecretory cells. Morphine 6-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 10648731-0 2000 Local morphine withdrawal increases c-fos gene, Fos protein, and oxytocin gene expression in hypothalamic magnocellular neurosecretory cells. Morphine 6-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 10648731-1 2000 We measured stimulation of c-fos and oxytocin gene expression during excitation of oxytocin cells induced by systemic or local morphine withdrawal. Morphine 127-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 10648731-3 2000 Morphine withdrawal increased the number of Fos-immunoreactive cells in the supraoptic and magnocellular paraventricular nuclei of conscious or pentobarbitone-anesthetized rats. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 10648731-3 2000 Morphine withdrawal increased the number of Fos-immunoreactive cells in the supraoptic and magnocellular paraventricular nuclei of conscious or pentobarbitone-anesthetized rats. Pentobarbital 144-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 10648731-4 2000 Morphine withdrawal also increased Fos-immunoreactive cell numbers in the parvocellular paraventricular nucleus of conscious but not anesthetized rats. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 10648731-6 2000 Unilateral microdialysis administration of naloxone (10(-5) M) into the supraoptic nucleus of anesthetized morphine-dependent rats increased Fos-immunoreactive cell numbers compared with the contralateral nucleus. Naloxone 43-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 10648731-6 2000 Unilateral microdialysis administration of naloxone (10(-5) M) into the supraoptic nucleus of anesthetized morphine-dependent rats increased Fos-immunoreactive cell numbers compared with the contralateral nucleus. Morphine 107-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 10648731-7 2000 Finally, we investigated whether dependence could be induced by chronic unilateral infusion of morphine into a supraoptic nucleus; systemic naloxone (5 mg/kg) increased Fos-immunoreactive cell numbers in the morphine-infused nucleus compared with the contralateral nucleus. Morphine 95-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-172 10648731-7 2000 Finally, we investigated whether dependence could be induced by chronic unilateral infusion of morphine into a supraoptic nucleus; systemic naloxone (5 mg/kg) increased Fos-immunoreactive cell numbers in the morphine-infused nucleus compared with the contralateral nucleus. Naloxone 140-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-172 10648731-7 2000 Finally, we investigated whether dependence could be induced by chronic unilateral infusion of morphine into a supraoptic nucleus; systemic naloxone (5 mg/kg) increased Fos-immunoreactive cell numbers in the morphine-infused nucleus compared with the contralateral nucleus. Morphine 208-216 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-172 10648731-8 2000 Thus, morphine withdrawal excitation increases c-fos and oxytocin gene expression in supraoptic nucleus neurons. Morphine 6-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 10776677-7 2000 These results show that (1) chronic alcohol consumption desensitizes or decreases the DAT of DA terminals in the ACC and that (2) EtOH causes cellular hyperexcitability of ACC dopaminergic neurons with increased Fos expression during alcohol tolerance. Alcohols 234-241 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 212-215 10764960-0 2000 L-prolyl-l-leucyl-glycinamide and its peptidomimetic analog 3(R)-[(2(S)-pyrrolidylcarbonyl)amino]-2-oxo-1-pyrrolidineacetamide (PAOPA) attenuate haloperidol-induced c-fos expression in the striatum. MSH Release-Inhibiting Hormone 0-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 10764960-0 2000 L-prolyl-l-leucyl-glycinamide and its peptidomimetic analog 3(R)-[(2(S)-pyrrolidylcarbonyl)amino]-2-oxo-1-pyrrolidineacetamide (PAOPA) attenuate haloperidol-induced c-fos expression in the striatum. 3(r)-[(2(s)-pyrrolidylcarbonyl)amino]-2-oxo-1-pyrrolidineacetamide 60-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 10764960-1 2000 Acute treatment of rats with haloperidol results in a rapid and transient increase in striatal c-fos mRNA and Fos immunoreactivity. Haloperidol 29-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 10764960-1 2000 Acute treatment of rats with haloperidol results in a rapid and transient increase in striatal c-fos mRNA and Fos immunoreactivity. Haloperidol 29-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 10764960-4 2000 We investigated the modulatory effects of PLG on haloperidol-induced c-fos and Fos protein expression in the rat striatum. Haloperidol 49-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 10800751-0 2000 A low dose of lithium chloride selectively induces Fos protein in the central nucleus of the amygdala of rat brain. Lithium Chloride 14-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 10764960-4 2000 We investigated the modulatory effects of PLG on haloperidol-induced c-fos and Fos protein expression in the rat striatum. Haloperidol 49-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 10800751-3 2000 To elucidate the neural substrates of the mood stabilizing actions of lithium, in the present study the authors investigated the effects of a low dose of lithium on regional expression of Fos protein. Lithium 154-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 188-191 10764960-5 2000 We report that coadministration of either PLG or the potent analog of PLG, 3(R)-[(2(S)-pyrrolidylcarbonyl)amino]-2-oxo-1-pyrrolidineacetam ide (PAOPA), attenuated haloperidol-induced c-fos and Fos expression. 3(r)-[(2(s)-pyrrolidylcarbonyl)amino]-2-oxo-1-pyrrolidineacetam ide 75-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 183-188 10800751-5 2000 The administrations of a high dose of lithium chloride (100 mg/kg) induced Fos in widespread areas of the rat brain. Lithium Chloride 38-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 10764960-5 2000 We report that coadministration of either PLG or the potent analog of PLG, 3(R)-[(2(S)-pyrrolidylcarbonyl)amino]-2-oxo-1-pyrrolidineacetam ide (PAOPA), attenuated haloperidol-induced c-fos and Fos expression. 3(r)-[(2(s)-pyrrolidylcarbonyl)amino]-2-oxo-1-pyrrolidineacetam ide 75-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-196 10627313-0 2000 Androgenic-anabolic steroids blunt morphine-induced c-fos expression in the rat striatum: possible role of beta-endorphin. Steroids 20-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 10784117-5 2000 Immunohistochemical assessment of c-fos expression also showed a difference between the two strains with preferential expression in the dorsal region of the rostral and caudal periaqueductal grey (PAG) in the hooded Lister rat, while the expression occurred in the ventral PAG in the Wistar rats. pag 197-200 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 10784117-5 2000 Immunohistochemical assessment of c-fos expression also showed a difference between the two strains with preferential expression in the dorsal region of the rostral and caudal periaqueductal grey (PAG) in the hooded Lister rat, while the expression occurred in the ventral PAG in the Wistar rats. pag 273-276 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 10627313-0 2000 Androgenic-anabolic steroids blunt morphine-induced c-fos expression in the rat striatum: possible role of beta-endorphin. Morphine 35-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 10627313-4 2000 In the present study, chronic administration of AAS blunted the striatal c-fos response to morphine, indicating that AAS can alter the molecular responses to at least one drug of abuse. aas 48-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 10627313-4 2000 In the present study, chronic administration of AAS blunted the striatal c-fos response to morphine, indicating that AAS can alter the molecular responses to at least one drug of abuse. Morphine 91-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 10627313-4 2000 In the present study, chronic administration of AAS blunted the striatal c-fos response to morphine, indicating that AAS can alter the molecular responses to at least one drug of abuse. aas 117-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 10627317-0 2000 NGF treatment potentiates c-fos expression in the rat nucleus basalis upon excitotoxic lesion with quisqualic acid. Quisqualic Acid 99-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 10627317-1 2000 The induction of the c-fos gene in the rat brain by NGF was studied in a model of acute cholinergic hypofunction, i.e., the lesion of the nucleus basalis magnocellularis (NBM) with quisqualic acid. Quisqualic Acid 181-196 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 10678749-1 2000 Systemic administration of the mixed 5-HT(1A/1B) agonist RU-24969 has been shown to produce a dramatic increase in locomotor activity and to induce robust c-Fos expression in the rat striatum. 5-methoxy 3-(1,2,3,6-tetrahydro-4-pyridinyl)1H indole 57-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-160 10728372-8 2000 ASMC and PAC-1 cells but not A10 showed a decrease in c-fos mRNA in response to 1 microgram/ml heparin, and a decrease in the c-Fos content of AP-1 DNA binding activity. Heparin 95-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 10728372-12 2000 In contrast to A10 and their controls not exposed to continuous heparin, heparin-selected PAC-1 and ASMC showed a diminished ability to induce c-fos in response to serum. Heparin 73-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-148 10728372-12 2000 In contrast to A10 and their controls not exposed to continuous heparin, heparin-selected PAC-1 and ASMC showed a diminished ability to induce c-fos in response to serum. asmc 100-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-148 10728372-13 2000 CONCLUSIONS: Smooth muscle cell lines show different responses to the antimitogenic effects of heparin that correlate with the heparin sensitivity of c-Fos/c-Jun expression. Heparin 95-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-155 10728372-13 2000 CONCLUSIONS: Smooth muscle cell lines show different responses to the antimitogenic effects of heparin that correlate with the heparin sensitivity of c-Fos/c-Jun expression. Heparin 127-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-155 10648884-0 2000 Calmodulin-dependent activation of p38 and p42/44 mitogen-activated protein kinases contributes to c-fos expression by calcium in PC12 cells: modulation by nitric oxide. Calcium 119-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 10648884-0 2000 Calmodulin-dependent activation of p38 and p42/44 mitogen-activated protein kinases contributes to c-fos expression by calcium in PC12 cells: modulation by nitric oxide. Nitric Oxide 156-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 10648884-2 2000 In the present study, we have investigated the roles of two mitogen-activated protein (MAP) kinases, extracellular signal-regulated protein kinase (ERK) and p38 MAP kinase (p38 kinase) in calcium- and NO-induced c-fos expression in PC12 cells. Calcium 188-195 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 212-217 10648884-5 2000 Calcium-induced c-fos expression was significantly reduced by the pretreatment of either MEK inhibitor (PD98059) or p38 kinase inhibitor (SB203580). Calcium 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 10648884-5 2000 Calcium-induced c-fos expression was significantly reduced by the pretreatment of either MEK inhibitor (PD98059) or p38 kinase inhibitor (SB203580). 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 104-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 10648884-5 2000 Calcium-induced c-fos expression was significantly reduced by the pretreatment of either MEK inhibitor (PD98059) or p38 kinase inhibitor (SB203580). SB 203580 138-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 10648884-6 2000 This finding indicates that the calmodulin-dependent activation of ERK and p38 kinase is involved in calcium-induced c-fos expression. Calcium 101-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 10648884-8 2000 NO-induced c-fos expression was partially inhibited by the PD98059. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 59-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 10648884-9 2000 We also observed that NO dose-dependently potentiates not only calcium-induced c-fos expression but also calcium-induced ERK activation. Calcium 63-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 10648884-10 2000 In the presence of PD98059, the amplification of calcium-induced c-fos expression by NO was not observed. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 19-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 10648884-10 2000 In the presence of PD98059, the amplification of calcium-induced c-fos expression by NO was not observed. Calcium 49-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 10686523-5 2000 In rats that were hemorrhaged on both D1 and D2, the responses of ACTH and corticosterone increased significantly from the first (H1) to the second hemorrhage (H2) in a control group injected with saline (p < 0.05) and in lesioned groups without bilateral damage of the Fos-responsive areas (p < 0.01). Sodium Chloride 197-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 273-276 10661499-0 2000 C-fos gene expression in rat brain around birth: effect of asphyxia and catecholamines. Catecholamines 72-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 10661499-11 2000 Therefore, we propose that while catecholamines play an important role in the induction of c-fos in the brain at birth, the effects of asphyxia involve a different mechanism. Catecholamines 33-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 10678749-2 2000 Previous studies have also shown that pretreatment with the D2-like dopamine agonist quinpirole virtually abolishes RU-24969-induced striatal c-Fos expression. Dopamine 68-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-147 10678749-2 2000 Previous studies have also shown that pretreatment with the D2-like dopamine agonist quinpirole virtually abolishes RU-24969-induced striatal c-Fos expression. Quinpirole 85-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-147 10678749-2 2000 Previous studies have also shown that pretreatment with the D2-like dopamine agonist quinpirole virtually abolishes RU-24969-induced striatal c-Fos expression. 5-methoxy 3-(1,2,3,6-tetrahydro-4-pyridinyl)1H indole 116-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-147 10678749-5 2000 RU-24969 alone produced elevations in locomotor activity and induced clear expression of c-Fos, FosB and Fra-2 throughout the entire striatal complex. 5-methoxy 3-(1,2,3,6-tetrahydro-4-pyridinyl)1H indole 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 11450029-6 2000 These results for the first time suggest that trauma induced release of serotonin and edema formation are important biological signals inducing c-fos expression. Serotonin 72-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-149 10849669-4 2000 Given the similarities between NO and O2, we hypothesized that NO inhibits hypoxia-induced up-regulation of c-fos and TH. Oxygen 38-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 10849669-7 2000 Hypoxia (1% O2 for 6 h) up-regulated c-fos and TH mRNA and increased c-fos promoter activity. Oxygen 12-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 10849669-7 2000 Hypoxia (1% O2 for 6 h) up-regulated c-fos and TH mRNA and increased c-fos promoter activity. Oxygen 12-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 11450029-0 2000 Spinal cord injury induced c-fos expression is reduced by p-CPA, a serotonin synthesis inhibitor. Fenclonine 58-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 11026470-0 2000 Margatoxin and iberiotoxin, two selective potassium channel inhibitors, induce c-fos like protein and mRNA in rat organotypic dorsal striatal slices. iberiotoxin 15-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 11450029-0 2000 Spinal cord injury induced c-fos expression is reduced by p-CPA, a serotonin synthesis inhibitor. Serotonin 67-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 11450029-2 2000 Influence of serotonin on upregulation of cellular-fos (c-fos) following a focal spinal cord injury was examined using immunohistochemistry in a rat model. Serotonin 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-54 11450029-2 2000 Influence of serotonin on upregulation of cellular-fos (c-fos) following a focal spinal cord injury was examined using immunohistochemistry in a rat model. Serotonin 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 11450029-5 2000 Pretreatment with p-CPA, a serotonin synthesis inhibitor, significantly attenuated the c-fos upregulation along with the edematous expansion of the cord. Fenclonine 18-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 11450029-5 2000 Pretreatment with p-CPA, a serotonin synthesis inhibitor, significantly attenuated the c-fos upregulation along with the edematous expansion of the cord. Serotonin 27-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 11026470-2 2000 In this study the effects of margatoxin and iberiotoxin on the expression of c-fos mRNA by in situ hybridization as well as on c-fos like protein by immunohistochemistry in isolated dorsal striatum after 10 days in culture were investigated. iberiotoxin 44-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 11026470-3 2000 C-fos mRNA dose-dependently increased 30 min after incubation with margatoxin and iberiotoxin. iberiotoxin 82-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 10902897-0 2000 Estradiol induces a phasic Fos response in the hippocampal CA1 and CA3 regions of adult female rats. Estradiol 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 11254161-7 2000 This association is suggested by transcriptional activation of the immediate-early response genes, c-fos and c-jun, within 15 min after exposure to cadmium and by the enhancement of AP-1 DNA binding activity, involving a c-Jun protein complex, which is maximally induced (approximately 4-fold) by 2 h. These molecular changes likely function together to protect alveolar epithelial cells against cadmium toxicity. Cadmium 148-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 11254161-7 2000 This association is suggested by transcriptional activation of the immediate-early response genes, c-fos and c-jun, within 15 min after exposure to cadmium and by the enhancement of AP-1 DNA binding activity, involving a c-Jun protein complex, which is maximally induced (approximately 4-fold) by 2 h. These molecular changes likely function together to protect alveolar epithelial cells against cadmium toxicity. Cadmium 396-403 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 10627287-0 2000 Essentiality of intron control in the induction of c-fos by glucose and glucoincretin peptides in INS-1 beta-cells. Glucose 60-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 10627287-2 2000 Such control is likely mediated via the expression of immediate-early response genes since several of these genes including c-fos are strongly induced by glucose in the beta-cell line INS-1, provided costimulation with cAMP-raising glucoincretin hormones. Glucose 154-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 10627287-2 2000 Such control is likely mediated via the expression of immediate-early response genes since several of these genes including c-fos are strongly induced by glucose in the beta-cell line INS-1, provided costimulation with cAMP-raising glucoincretin hormones. Cyclic AMP 219-223 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 10627287-3 2000 This study addresses the mechanism of c-fos gene activation by glucose. Glucose 63-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 10627287-4 2000 Glucose in the presence of chlorophenylthio-cAMP generated a low threefold induction of the c-fos/basic luciferase reporter gene, which includes only the c-fos promoter. Glucose 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 10627287-4 2000 Glucose in the presence of chlorophenylthio-cAMP generated a low threefold induction of the c-fos/basic luciferase reporter gene, which includes only the c-fos promoter. Glucose 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 10627287-4 2000 Glucose in the presence of chlorophenylthio-cAMP generated a low threefold induction of the c-fos/basic luciferase reporter gene, which includes only the c-fos promoter. 8-(4-Chlorophenylthio)-cAMP 27-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 10627287-4 2000 Glucose in the presence of chlorophenylthio-cAMP generated a low threefold induction of the c-fos/basic luciferase reporter gene, which includes only the c-fos promoter. 8-(4-Chlorophenylthio)-cAMP 27-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 10627287-11 2000 In conclusion, the strong induction of c-fos by glucose and glucoincretins results from Ca(2+) and cAMP signaling pathways addressing both the CRE in the promoter and essential response element(s) in the first intron that are unrelated to the transcription arrest site. Glucose 48-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 10627287-11 2000 In conclusion, the strong induction of c-fos by glucose and glucoincretins results from Ca(2+) and cAMP signaling pathways addressing both the CRE in the promoter and essential response element(s) in the first intron that are unrelated to the transcription arrest site. Cyclic AMP 99-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 10902897-7 2000 Our results show a phasic estradiol-induced c-Fos response in the pyramidal cell layers of both CA1 and CA3. Estradiol 26-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 10902897-8 2000 c-Fos was induced within 2 h of treatment, decreased at 6 and 12 h, and subsequently increased again at 24 h after treatment with estradiol. Estradiol 130-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 10902897-10 2000 These estradiol-induced changes in c-Fos expression may reflect phasic neuronal activation and coupling to gene expression, which could be involved in estradiol"s effects on excitatory synaptic connectivity in the hippocampus. Estradiol 6-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 10854763-11 2000 The combined application of N-methyl-D-aspartate 340 microM + substance P (at 0.74 or 3.7 microM) significantly increased ipsilateral c-Fos compared to either agent alone. N-Methylaspartate 28-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-139 10642317-8 2000 The specific Rho-kinase inhibitor, Y-27632, dose-dependently abolished Ang II-induced protein synthesis and also suppressed Ang II-induced c-fos mRNA expression. Y 27632 35-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 10567852-8 2000 Competitive reverse transcription-polymerase chain reaction showed a high level of glomerular c-fos mRNA in high-salt-loaded rats and that ACEI or AT1a treatment did not significantly change its level. Salts 113-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 10854763-0 2000 Simultaneous activation of N-methyl-D-aspartate and neurokinin-1 receptors modulates c-Fos and Zif/268 expression in the rat trigeminal nucleus caudalis. Aspartic Acid 37-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 10854764-1 2000 We examined the effects of systemic administration of a GABA(A) receptor agonist, muscimol, or antagonist, bicuculline, on the expression of c-Fos protein induced 3h after electrical stimulation of the trigeminal ganglion at low (0.1 mA) or high intensities (1. gamma-Aminobutyric Acid 56-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 10854763-4 2000 Ipsilateral c-Fos and Zif/268 were increased significantly dose-dependently by N-methyl-D-aspartate (at 136 and 340 microM, and at 68, 136 and 340 microM, respectively). N-Methylaspartate 79-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 10854764-1 2000 We examined the effects of systemic administration of a GABA(A) receptor agonist, muscimol, or antagonist, bicuculline, on the expression of c-Fos protein induced 3h after electrical stimulation of the trigeminal ganglion at low (0.1 mA) or high intensities (1. Muscimol 82-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 10854764-1 2000 We examined the effects of systemic administration of a GABA(A) receptor agonist, muscimol, or antagonist, bicuculline, on the expression of c-Fos protein induced 3h after electrical stimulation of the trigeminal ganglion at low (0.1 mA) or high intensities (1. Bicuculline 107-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 10854764-1 2000 We examined the effects of systemic administration of a GABA(A) receptor agonist, muscimol, or antagonist, bicuculline, on the expression of c-Fos protein induced 3h after electrical stimulation of the trigeminal ganglion at low (0.1 mA) or high intensities (1. Tritium 163-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 10854764-3 2000 In saline-treated rats, 10 min stimulation of the trigeminal ganglion induced c-Fos-immunopositive neurons throughout the full extent of the ipsilateral superficial layers of the trigeminal nucleus caudalis, and dorsal or dorsomedial part of the nuclei rostral to obex (trigeminal nucleus principalis, dorsomedial nucleus of trigeminal nucleus oralis, dorsomedial nucleus of trigeminal nucleus interpolaris). Sodium Chloride 3-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 10854764-7 2000 muscimol and stimulated at 0.1 mA, the numbers of Fos-positive neurons in trigeminal nucleus caudalis, dorsomedial nucleus of trigeminal nucleus interpolaris, and dorsomedial nucleus of trigeminal nucleus oralis were significantly decreased. Muscimol 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 11068145-2 2000 Transganglionic transport and denervation methods were used in an experimental setting to test the dependence of phenylephrine-induced Fos immunoreactivity on the integrity of buffer nerve afferents, and to identify the subregions of the nucleus of the solitary tract supplied by each. Phenylephrine 113-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-138 10854764-10 2000 bicuculline and stimulated at 0.1 mA, the number of Fos-positive neurons increased in the superficial layers of trigeminal nucleus caudalis and trigeminal nucleus principalis. Bicuculline 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 11068145-8 2000 Animals killed 30 days after complete sinoaortic denervation displayed no evidence of recovery of phenylephrine-induced Fos, while the strength and distribution of the response in rats that received selective carotid sinus denervation were indistinguishable from those seen in controls. Phenylephrine 98-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-123 10858613-2 2000 In addition, we found an inverse correlation between c-Fos protein and CArG-binding protein A messenger RNA levels in the lateral caudate-putamen of rats treated acutely and chronically with the D2 receptor antagonist fluphenazine (phenothiozine typical psychotic). Fluphenazine 218-230 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 11068145-9 2000 These findings (i) support the dependence of phenylephrine-induced Fos expression on the integrity of carotid sinus and aortic depressor nerve afferents, (ii) provide anatomical and functional evidence that the two buffer nerves distribute differentially within the nucleus of the solitary tract, and (iii) implicate central reorganization as a likely basis for functional recovery of baroreflex mechanisms following partial sinoaortic denervation. Phenylephrine 45-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 10858613-2 2000 In addition, we found an inverse correlation between c-Fos protein and CArG-binding protein A messenger RNA levels in the lateral caudate-putamen of rats treated acutely and chronically with the D2 receptor antagonist fluphenazine (phenothiozine typical psychotic). phenothiozine 232-245 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 10858623-4 2000 The number of neurons expressing Fos, determined at the end of the fifth day, was increased in the adrenalectomized rats with or without NaCl solution to drink. nacl solution 137-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 10858623-6 2000 Treatment of adrenalectomized rats with the sodium-retaining mineralocorticoid hormone, deoxycorticosterone, at the end of the fourth day, decreased Fos expression in the subfornical organ and the organum vasculosum of the lamina terminalis when NaCl solution was available but not when the NaCl solution was unavailable. Sodium 44-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 149-152 10858623-6 2000 Treatment of adrenalectomized rats with the sodium-retaining mineralocorticoid hormone, deoxycorticosterone, at the end of the fourth day, decreased Fos expression in the subfornical organ and the organum vasculosum of the lamina terminalis when NaCl solution was available but not when the NaCl solution was unavailable. Desoxycorticosterone 88-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 149-152 10858623-9 2000 Relative to sodium-deplete intact rats, however, sodium-deplete adrenalectomized rats had a greater number of neurons expressing Fos in the organum vasculosum. Sodium 49-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-132 10858623-10 2000 Treatment of sodium-deplete rats, adrenalectomized or intact, with the angiotensin II-type 1 receptor antagonist, ZD7155, decreased sodium intake and Fos expression in the subfornical organ but not in the organum vasculosum of the lamina terminalis or median preoptic nucleus. Sodium 13-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-153 10619471-1 2000 We investigated the role of calcium/calmodulin-dependent protein kinases in the phosphorylation of cyclic AMP response element binding protein and subsequent induction of c-fos gene elicited by sustained hypertension in neurons of the nucleus tractus solitarii of anesthetized rats. Cyclic AMP 99-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-176 10658625-0 2000 c-fos expression, behavioural, endocrine and autonomic responses to acute social stress in male rats after chronic restraint: modulation by serotonin. Serotonin 140-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 10619471-7 2000 The present results suggest that activation of calcium/calmodulin-dependent protein kinases may represent an important step in the cascade of intracellular events that leads to phosphorylation of cyclic AMP response element binding protein and subsequent induction of c-fos gene after activation of ionotropic glutamate receptors by baroceptive signals in the nucleus tractus solitarii. Cyclic AMP 196-206 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 268-273 10658620-3 2000 Previous results have shown that unilateral stimulation of metabotropic glutamate receptors in the subthalamic nucleus with the non-subtype-selective metabotropic glutamate receptor agonist 1S,3R-1-amino-1,3-cyclopentane dicarboxylate results in contralateral rotation in rats and Fos expression in the subthalamic nucleus. 1s 190-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 281-284 10658625-7 2000 Serotonin depletion reduced the expression of c-fos in the frontal cortex, lateral preoptic area, medial amygdala, central gray, medial and dorsal raphe, and locus coeruleus after social stress, but this was not altered by previous restraint. Serotonin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 10658633-5 2000 This was accompanied by a marked reduction in formalin-induced c-fos expression in the spinal dorsal horn. Formaldehyde 46-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 10658620-3 2000 Previous results have shown that unilateral stimulation of metabotropic glutamate receptors in the subthalamic nucleus with the non-subtype-selective metabotropic glutamate receptor agonist 1S,3R-1-amino-1,3-cyclopentane dicarboxylate results in contralateral rotation in rats and Fos expression in the subthalamic nucleus. 3r-1-amino-1,3-cyclopentane dicarboxylate 193-234 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 281-284 10682714-0 2000 Lidocaine blockade of amygdala output in fear-conditioned rats reduces Fos expression in the ventrolateral periaqueductal gray. Lidocaine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 10670444-9 2000 Most of the Fos-labeled cells co-expressed pro-thyrotropin-releasing hormone messenger RNA signal and/or were serotonin immunoreactive. Serotonin 110-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-15 10670449-0 2000 Tachykinin receptor inhibition and c-Fos expression in the rat brain following formalin-induced pain. Formaldehyde 79-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 10670449-2 2000 The expression of the inducible transcription factor, c-Fos, was used to identify areas in the brain activated by a noxious stimulus (the subcutaneous injection of formalin), and to investigate the effects of intracerebroventricular administration of selective, nonpeptide antagonists for neurokinin-1 and neurokinin-2 tachykinin receptors on the neural activity in these areas and on the behavioural response to formalin-induced pain. Formaldehyde 164-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 10670449-2 2000 The expression of the inducible transcription factor, c-Fos, was used to identify areas in the brain activated by a noxious stimulus (the subcutaneous injection of formalin), and to investigate the effects of intracerebroventricular administration of selective, nonpeptide antagonists for neurokinin-1 and neurokinin-2 tachykinin receptors on the neural activity in these areas and on the behavioural response to formalin-induced pain. Formaldehyde 413-421 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 10682714-8 2000 The results show that, when lidocaine was microinjected in the medial part of the central nucleus of the amygdala or along the ventral amygdalofugal pathway of conditioned rats, fear-induced Fos expression in the ventrolateral periaqueductal gray was reduced on the side ipsilateral to the injection (up to 37% reduction in comparison to the contralateral side). Lidocaine 28-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 191-194 10682714-10 2000 Fos expression remained low on both sides in the non-fear-conditioned animals injected with lidocaine. Lidocaine 92-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 10682714-11 2000 Finally, although freezing was only partly reduced in the conditioned animals unilaterally injected with lidocaine, it was significantly correlated to the ipsilateral reduction in Fos expression. Lidocaine 105-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 180-183 10683409-4 2000 Following 6-hydroxydopamine infusions into the medial forebrain bundle in awake, behaving rats, there was a rapid and transient induction of striatal c-fos and zif/268 messenger RNAs. Oxidopamine 10-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-155 10670449-3 2000 Formalin (5%, 50 microl), injected subcutaneously through a chronically implanted catheter in the region of the lower hindlimb, increased c-Fos expression in a number of brain areas related to nociceptive transmission or the integration of stress responses. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 10670449-6 2000 The neurokinin-1, but not the neurokinin-2, receptor antagonist attenuated the formalin-induced activation of c-Fos in the paraventricular nucleus of the hypothalamus. Formaldehyde 79-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 10683409-7 2000 6-Hydroxydopamine-induced c-fos messenger RNA was also observed in the substantia nigra pars reticulata and entopeduncular nucleus, but not the globus pallidus, 45 min after medial forebrain bundle 6-hydroxydopamine infusions. Oxidopamine 0-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 10683575-0 2000 Fos expression is induced by increased nitric oxide release in rat spinal cord dorsal horn. Nitric Oxide 39-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 10683409-7 2000 6-Hydroxydopamine-induced c-fos messenger RNA was also observed in the substantia nigra pars reticulata and entopeduncular nucleus, but not the globus pallidus, 45 min after medial forebrain bundle 6-hydroxydopamine infusions. Oxidopamine 198-215 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 10683575-1 2000 The relationship between exogenous or endogenous nitric oxide and c-fos, an immediate-early gene which can further activate the production of other substances in the central nervous system, was investigated in this study. Nitric Oxide 49-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 10683409-9 2000 Both the N-methyl-D-aspartate antagonist, (+/-)-3-(2-carboxypiperazin-4-yl)-propyl-1-phosphonic acid, and the non-N-methyl-D-aspartate antagonist, 6,7-dinitroquinoxaline-2, 3-dione, blocked striatal induction of c-fos messenger RNA following 6-hydroxydopamine infusions into the medial forebrain bundle. 3-(2-carboxypiperazin-4-yl)propyl-1-phosphonic acid 42-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 212-217 10683575-2 2000 We found that Fos expression is increased after intradermal capsaicin injection, which also leads to endogenous nitric oxide release in the spinal cord. Capsaicin 60-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 10683575-2 2000 We found that Fos expression is increased after intradermal capsaicin injection, which also leads to endogenous nitric oxide release in the spinal cord. Nitric Oxide 112-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 10828527-1 2000 In this study we describe the preventive effect of interruption of the supramammillohippocampal afferents on the Fos expression in the forebrain and epileptic discharges in the hippocampal electroencephalogram in rat model of kainic acid-induced limbic seizure. Kainic Acid 226-237 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-116 10683575-4 2000 The increased Fos expression is blocked by N(G)-nitro-L-arginine methyl ester, a nitric oxide synthase inhibitor, but not by its inactive isomer N(G)-nitro-D-arginine methyl ester. NG-Nitroarginine Methyl Ester 43-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 10683575-7 2000 Although Fos expression was increased with 3-morpholino-sydnonimine perfusion compared to that seen with artificial cerebrospinal fluid perfusion, there was still some Fos immunostaining in the control sections. linsidomine 43-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 10683575-8 2000 Following perfusion of artificial cerebrospinal fluid in the spinal cord of rats pretreated with N(G)-nitro-L-arginine methyl ester, it was found that Fos staining was reduced significantly compared to the control sections from animals without N(G)-nitro-L-arginine methyl ester pretreatment. NG-Nitroarginine Methyl Ester 97-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-154 10683575-8 2000 Following perfusion of artificial cerebrospinal fluid in the spinal cord of rats pretreated with N(G)-nitro-L-arginine methyl ester, it was found that Fos staining was reduced significantly compared to the control sections from animals without N(G)-nitro-L-arginine methyl ester pretreatment. NG-Nitroarginine Methyl Ester 244-278 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-154 10683575-9 2000 These results suggest that nitric oxide helps mediate Fos expression induced by an intradermal capsaicin injection. Nitric Oxide 27-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 10683575-9 2000 These results suggest that nitric oxide helps mediate Fos expression induced by an intradermal capsaicin injection. Capsaicin 95-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 10683575-10 2000 We conclude that both endogenous and exogenous nitric oxide induce Fos expression. Nitric Oxide 47-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 10683575-11 2000 Involvement of nitric oxide in the development of central sensitization may affect nociceptive processing by increasing Fos expression. Nitric Oxide 15-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-123 10683575-12 2000 Since many other substances which are related to pain mechanisms can be induced by Fos, it is suggested that nitric oxide may regulate production of these substances through activation of Fos. Nitric Oxide 109-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 10828527-6 2000 Muscimol injection to the supramammillary nucleus prevented Fos expression in the CA1-CA3 region and reduced that in the forebrain regions with the latest Fos expression, but did not affect Fos expression in other forebrain regions with early Fos expression. Muscimol 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 10683575-12 2000 Since many other substances which are related to pain mechanisms can be induced by Fos, it is suggested that nitric oxide may regulate production of these substances through activation of Fos. Nitric Oxide 109-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 188-191 10828527-6 2000 Muscimol injection to the supramammillary nucleus prevented Fos expression in the CA1-CA3 region and reduced that in the forebrain regions with the latest Fos expression, but did not affect Fos expression in other forebrain regions with early Fos expression. Muscimol 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-158 10828527-6 2000 Muscimol injection to the supramammillary nucleus prevented Fos expression in the CA1-CA3 region and reduced that in the forebrain regions with the latest Fos expression, but did not affect Fos expression in other forebrain regions with early Fos expression. Muscimol 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-158 10828527-6 2000 Muscimol injection to the supramammillary nucleus prevented Fos expression in the CA1-CA3 region and reduced that in the forebrain regions with the latest Fos expression, but did not affect Fos expression in other forebrain regions with early Fos expression. Muscimol 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-158 10579804-0 2000 Selective c-fos induction and decreased dopamine release in the central nucleus of amygdala in rats displaying a mecamylamine-precipitated nicotine withdrawal syndrome. Mecamylamine 113-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-15 10994593-5 2000 In barbiturate-anesthetized male rats electrical stimulation of XIIn produced a significant increase in Fos-positive neurons in the dorsal paratrigeminal nucleus (dPa5) and laminae I-II of caudal subnucleus caudalis (Vc) and upper cervical dorsal horn. barbituric acid 3-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 10994593-6 2000 Mustard oil injection into the deep tongue muscle also produced an increase in c-fos expression in dPa5; however, the highest density of expression occurred in laminae I-II at the dorsomedial aspect of rostral Vc. mustard oil 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 10579804-0 2000 Selective c-fos induction and decreased dopamine release in the central nucleus of amygdala in rats displaying a mecamylamine-precipitated nicotine withdrawal syndrome. Nicotine 139-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-15 10579804-1 2000 In the present study the neuronal expression of Fos, the protein product of c-fos, was used to study changes in neuronal activity in nerve terminal regions of the ascending dopaminergic system during nicotine withdrawal. Nicotine 200-208 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 10579804-1 2000 In the present study the neuronal expression of Fos, the protein product of c-fos, was used to study changes in neuronal activity in nerve terminal regions of the ascending dopaminergic system during nicotine withdrawal. Nicotine 200-208 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 10579804-6 2000 The number of Fos-positive nuclei was substantially increased in the central nucleus of amygdala (CNA) in animals undergoing mecamylamine-precipitated withdrawal, whereas no significant changes in c-fos expression were observed in the basolateral amygdaloid nucleus, the core and the shell of the nucleus accumbens, the dorsolateral striatum, or the medial prefrontal cortex. Mecamylamine 125-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 10716222-4 1999 After hemorrhage most Fos-positive catecholamine cells were found below obex whereas most hypoxia-responsive cells were rostral to obex. Catecholamines 35-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-25 10642850-2 1999 Using dual-immunocytochemistry, the number of oxytocinergic neurons in the hypothalamic magnocellular nuclei co-storing Fos after administration of D-fenfluramine was found to be 15-fold higher compared to vehicle, comprising about 30% of the total number of these neurons. Dexfenfluramine 148-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-123 10637445-8 1999 Striatal AdGDNF injections also reduced tyrosine hydroxylase fiber loss and increased amphetamine-induced striatal Fos expression. Amphetamine 86-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-118 10598624-8 1999 The expression of c-Fos induced by potassium chloride was also suppressed by dibucaine, tetracaine (concentrations that inhibited 50% of the activity of positive control [IC50s] were 16.2+/-0.2 and 73.2+/-0.7 microM, respectively), and PD 98059, a mitogen-activated/extracellular receptor-regulated kinase inhibitor. Potassium Chloride 35-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 10598624-8 1999 The expression of c-Fos induced by potassium chloride was also suppressed by dibucaine, tetracaine (concentrations that inhibited 50% of the activity of positive control [IC50s] were 16.2+/-0.2 and 73.2+/-0.7 microM, respectively), and PD 98059, a mitogen-activated/extracellular receptor-regulated kinase inhibitor. Dibucaine 77-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 10598624-8 1999 The expression of c-Fos induced by potassium chloride was also suppressed by dibucaine, tetracaine (concentrations that inhibited 50% of the activity of positive control [IC50s] were 16.2+/-0.2 and 73.2+/-0.7 microM, respectively), and PD 98059, a mitogen-activated/extracellular receptor-regulated kinase inhibitor. Tetracaine 88-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 10598624-11 1999 CONCLUSIONS: Dibucaine and tetracaine at clinical concentrations were found to inhibit the activation of MAP kinase and the expression of c-Fos mediated by L-type Ca2+ channels in PC12 cells. Dibucaine 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 10598624-11 1999 CONCLUSIONS: Dibucaine and tetracaine at clinical concentrations were found to inhibit the activation of MAP kinase and the expression of c-Fos mediated by L-type Ca2+ channels in PC12 cells. Tetracaine 27-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 10594656-4 1999 In freely moving rats, we stimulated the frontal cortex by local epidural application of a dose of a GABAA receptor antagonist (picrotoxin) just threshold for inducing Fos in the striatum. Picrotoxin 128-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-171 10567406-11 1999 Increased activation of these MAP kinase pathways may be one mechanism by which cGMP and PKG activation mediate c-fos induction and increased proliferation of contractile adult RASMC. Cyclic GMP 80-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 10657527-2 1999 In this study, we examined the effects of PACAP on the expression of fos and jun immediate early gene (IEG) families, expression of which can be up-regulated by both PKC and cAMP signaling pathways, in rat pheochromocytoma cell line PC12 cells. Cyclic AMP 174-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 10891619-9 2000 In summary, each of the anxiogenic drugs tested (FG-7142, yohimbine, m-chlorophenyl piperazine, caffeine and BOC-CCK(4)) increased Fos expression in a restricted number of hindbrain regions, including the periaqueductal gray and locus coeruleus. Yohimbine 58-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-134 10891619-9 2000 In summary, each of the anxiogenic drugs tested (FG-7142, yohimbine, m-chlorophenyl piperazine, caffeine and BOC-CCK(4)) increased Fos expression in a restricted number of hindbrain regions, including the periaqueductal gray and locus coeruleus. 1-(3-chlorophenyl)piperazine 69-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-134 10891619-9 2000 In summary, each of the anxiogenic drugs tested (FG-7142, yohimbine, m-chlorophenyl piperazine, caffeine and BOC-CCK(4)) increased Fos expression in a restricted number of hindbrain regions, including the periaqueductal gray and locus coeruleus. Caffeine 96-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-134 10593203-0 1999 Effects of formalin pain on hippocampal c-Fos expression in male and female rats. Formaldehyde 11-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 10575103-8 1999 Baclofen inhibited the Fos-LI expression both in the spinal cord and the gracile nucleus. Baclofen 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 10575103-9 1999 Morphine inhibited only the Fos-LI expression in the posterior cutaneous (PC) nerve territory of laminae I-II, but not in the sciatic nerve (SC) territory, laminae III-IV nor the gracile nucleus. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 10611432-9 1999 An ipsilateral administration of 1 micromol 5"-amino-5"-deoxyadenosine, but not 1 micromol 2"-deoxycoformycin, reduced carrageenan-induced c-Fos expression in the spinal dorsal horn, and this was further reduced by the peripheral co-injection of the two agents. 5'-amino-5'-deoxyadenosine 44-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 10593203-6 1999 In males at time 2, formalin increased c-Fos in the dentate gyrus (DG) and CA3 fields; at time 24, c-Fos returned to the control level; at time 24/OF, c-Fos was higher than in control in the DG, but not in the other fields. Formaldehyde 20-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 10593203-7 1999 In the formalin-treated females at time 2 and at time 24, c-Fos levels were lower, or tended to be lower, than in control in all hippocampal fields; at time 24/OF, c-Fos levels in the DG were higher than in control and in males. Formaldehyde 7-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 10593203-7 1999 In the formalin-treated females at time 2 and at time 24, c-Fos levels were lower, or tended to be lower, than in control in all hippocampal fields; at time 24/OF, c-Fos levels in the DG were higher than in control and in males. Formaldehyde 7-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-169 10605821-5 1999 It has also been demonstrated that spermidine, preferentially stimulated the transcription and the expression of c-myc while those of c-fos were preferentially stimulated by putrescine. Putrescine 174-184 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-139 10580588-6 1999 DMSO down-regulated c-myc and c-fos expression. Dimethyl Sulfoxide 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 10531466-7 1999 Morphine significantly reduced noxious stimulus-induced Fos expression in lamina I, but the Fos inhibition was less pronounced in neurons that expressed the NK-1 receptor. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 10587093-0 1999 Baclofen and midazolam alter c-fos induction by peripheral noxious or innocuous stimulation in the spinal cord of normal and monoarthritic rats. Baclofen 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 10587093-0 1999 Baclofen and midazolam alter c-fos induction by peripheral noxious or innocuous stimulation in the spinal cord of normal and monoarthritic rats. Midazolam 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 10587093-3 1999 caused a significant reduction in the number of Fos-positive neurones following noxious stimulation of both normal and monoarthritic animals, which was prevented by the GABA(B) antagonist CGP 35348 (200 mg/kg, i.v.). gamma-Aminobutyric Acid 169-173 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 10587093-3 1999 caused a significant reduction in the number of Fos-positive neurones following noxious stimulation of both normal and monoarthritic animals, which was prevented by the GABA(B) antagonist CGP 35348 (200 mg/kg, i.v.). CGP 35348 188-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 10587093-6 1999 Baclofen also reduced the number of Fos-positive neurones in monoarthritic animals subject to innocuous stimulation. Baclofen 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-39 10587093-11 1999 It can be concluded that although GABA(B) receptors modulate sensory input at the spinal level, high doses of systemic baclofen are required to inhibit nociceptive-induced c-fos expression. Baclofen 119-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-177 10587093-12 1999 The paradoxical facilitation of c-fos expression by midazolam in monoarthritic animals, may be due to the reported increase in spinal GABA levels found in those animals. Midazolam 52-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 10587093-12 1999 The paradoxical facilitation of c-fos expression by midazolam in monoarthritic animals, may be due to the reported increase in spinal GABA levels found in those animals. gamma-Aminobutyric Acid 134-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 11216448-0 1999 Intrathecal cannabinoid administration suppresses noxious stimulus-evoked Fos protein-like immunoreactivity in rat spinal cord: comparison with morphine. Cannabinoids 12-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 11216448-1 1999 AIM: To determine whether cannabinoids suppress noxious stimulus-evoked Fos protein-like immunoreactivity (FLI) through direct actions at the spinal level. Cannabinoids 26-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 11216448-5 1999 The cannabinoid suppressed Fos in the neck region of the dorsal horn and in the ventral horn, but not in the nucleus proprius. Cannabinoids 4-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 10533047-7 1999 Thus, blockade, under basal conditions, of the effect of spontaneously released dopamine by the D2 receptor antagonist haloperidol leads to the activation of c-Fos expression in the striatum. Dopamine 80-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 10533047-7 1999 Thus, blockade, under basal conditions, of the effect of spontaneously released dopamine by the D2 receptor antagonist haloperidol leads to the activation of c-Fos expression in the striatum. Haloperidol 119-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 10533047-8 1999 Furthermore, stimulation of DA release by amphetamine induces striatal c-Fos expression in a D1 receptor-dependent manner. Dopamine 28-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 10533047-8 1999 Furthermore, stimulation of DA release by amphetamine induces striatal c-Fos expression in a D1 receptor-dependent manner. Amphetamine 42-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 10581392-0 1999 Adenosine and behavioral state control: adenosine increases c-Fos protein and AP1 binding in basal forebrain of rats. Adenosine 40-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 10583477-8 1999 By using in situ hybridization it was found that CPA (0.1 mg/kg) significantly inhibited the SKF 38393-induced increase in the expression of NGFI-A and c-fos mRNA levels in the dopamine-denervated striatum. N(6)-cyclopentyladenosine 49-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-157 10583477-8 1999 By using in situ hybridization it was found that CPA (0.1 mg/kg) significantly inhibited the SKF 38393-induced increase in the expression of NGFI-A and c-fos mRNA levels in the dopamine-denervated striatum. Dopamine 177-185 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-157 10534582-7 1999 DELT given i.c.v., i.th., or into the MRF also blocked formalin-induced increase in Fos-like immunoreactivity (FLI) in the dorsal horn of lumbar spinal cord ipsilateral to the formalin injection. Formaldehyde 55-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 10534605-0 1999 The NMDA receptor antagonist MK-801 attenuates c-Fos expression in the lumbosacral spinal cord following repetitive noxious and non-noxious colorectal distention. Dizocilpine Maleate 29-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 10534605-1 1999 The effects of pretreatment with an NMDA receptor antagonist, MK-801, on c-Fos (Fos) expression in the lumbosacral spinal cord following repetitive, noxious (80 mmHg) or non-noxious (20 mmHg) colorectal distention (CRD) was examined immunocytochemically in awake and urethane anesthetized rats. Dizocilpine Maleate 62-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 10534605-9 1999 Five mg/kg MK-801 attenuated non-noxious CRD-induced Fos by 20%. Dizocilpine Maleate 11-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 10534605-11 1999 The laminar distribution of Fos following MK-801 pretreatment revealed a tendency towards the deeper laminae showing the greatest attenuation at the highest dose of MK-801. Dizocilpine Maleate 42-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 10534605-11 1999 The laminar distribution of Fos following MK-801 pretreatment revealed a tendency towards the deeper laminae showing the greatest attenuation at the highest dose of MK-801. Dizocilpine Maleate 165-171 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 10611432-9 1999 An ipsilateral administration of 1 micromol 5"-amino-5"-deoxyadenosine, but not 1 micromol 2"-deoxycoformycin, reduced carrageenan-induced c-Fos expression in the spinal dorsal horn, and this was further reduced by the peripheral co-injection of the two agents. Carrageenan 119-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 10585535-4 1999 Supershift analysis revealed that the carbachol-induced AP-1 complex was composed predominantly of Jun D and c-Fos. Carbachol 38-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 10516208-0 1999 Asbestos-induced phosphorylation of epidermal growth factor receptor is linked to c-fos and apoptosis. Asbestos 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 10502313-2 1999 Administration of 8-OH-DPAT (0.1 and 1 mg/kg) induced a marked and dose-related increase in the number of cells positive for Fos-LI in the locus coeruleus (LC), the main source of noradrenergic projections to the forebrain. 8-Hydroxy-2-(di-n-propylamino)tetralin 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-128 10502313-4 1999 The effect of both 8-OH-DPAT (0.1 mg/kg) and buspirone (10 mg/kg) on Fos-LI in the LC was blocked by pretreatment with the selective 5-HT(1A) receptor antagonist WAY 100635 (1 mg/kg). 8-Hydroxy-2-(di-n-propylamino)tetralin 19-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 10502313-4 1999 The effect of both 8-OH-DPAT (0.1 mg/kg) and buspirone (10 mg/kg) on Fos-LI in the LC was blocked by pretreatment with the selective 5-HT(1A) receptor antagonist WAY 100635 (1 mg/kg). Buspirone 45-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 10574568-3 1999 Previous work from this laboratory demonstrated that systemic administrations of the serotonin agonist quipazine mimic the effects of light on the circadian system of rats, i.e. they induce photic-like phase shifts of the circadian activity rhythm as well as c-Fos expression in the SCN. Serotonin 85-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 261-264 10574568-3 1999 Previous work from this laboratory demonstrated that systemic administrations of the serotonin agonist quipazine mimic the effects of light on the circadian system of rats, i.e. they induce photic-like phase shifts of the circadian activity rhythm as well as c-Fos expression in the SCN. Quipazine 103-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 261-264 10529449-0 1999 MK-801 administration blocks the effects of a 5-HT(2A/2C) agonist on melatonin rhythmicity and c-fos induction in the suprachiasmatic nucleus. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 10529449-2 1999 This study investigated the effects of the NMDA receptor antagonist, MK-801 on the phase shift of the melatonin rhythm and the induction of c-fos in the rat suprachiasmatic nucleus (SCN) provoked by the administration of the serotonin agonist DOI ((+/-)-1-(4-Iodo-2,5-dimethoxyphenyl)-2-aminopropane hydrochloride). Dizocilpine Maleate 69-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 10529449-2 1999 This study investigated the effects of the NMDA receptor antagonist, MK-801 on the phase shift of the melatonin rhythm and the induction of c-fos in the rat suprachiasmatic nucleus (SCN) provoked by the administration of the serotonin agonist DOI ((+/-)-1-(4-Iodo-2,5-dimethoxyphenyl)-2-aminopropane hydrochloride). Serotonin 225-234 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 10529449-5 1999 Pre-treatment with MK-801 also inhibited by approximately 90% the induction of c-fos in the SCN by DOI at ZT18 (6 h after actual darkness onset) as determined by immunohistochemistry. Dizocilpine Maleate 19-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 10516208-5 1999 The tyrphostin AG-1478, which inhibits the tyrosine kinase activity of the EGFR, but not the tyrphostin A-10, which does not affect EGFR activity, significantly ameliorated asbestos-induced increases in mRNA levels of c-fos but not of c-jun. Tyrphostins 4-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 218-223 10516208-7 1999 Our findings suggest that asbestos-induced binding to EGFR initiates signaling pathways responsible for increased expression of the protooncogene c-fos and the development of apoptosis. Asbestos 26-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-151 10516251-4 1999 In the dorsomedial, commissural, and gelatinosus subnuclei, MK-801 in itself produced significant Fos expression and significantly reduced (-75%, P < 0.05) the ability of gastric distension to induce Fos expression, assuming an additive model with two separate populations of neurons activated by distension and the blocker. Dizocilpine Maleate 60-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 10516251-4 1999 In the dorsomedial, commissural, and gelatinosus subnuclei, MK-801 in itself produced significant Fos expression and significantly reduced (-75%, P < 0.05) the ability of gastric distension to induce Fos expression, assuming an additive model with two separate populations of neurons activated by distension and the blocker. Dizocilpine Maleate 60-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 203-206 10512366-0 1999 Palmitate and oleate induce the immediate-early response genes c-fos and nur-77 in the pancreatic beta-cell line INS-1. Palmitates 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 10514457-3 1999 Blockade of calcium influx through nifedipine-sensitive voltage-gated calcium channels reduced buserelin-induced activation of extracellular signal-regulated kinase (ERK) and c-Fos while activation of c-Jun N-terminal kinase and c-Jun was unaffected. Calcium 12-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 175-180 10514457-3 1999 Blockade of calcium influx through nifedipine-sensitive voltage-gated calcium channels reduced buserelin-induced activation of extracellular signal-regulated kinase (ERK) and c-Fos while activation of c-Jun N-terminal kinase and c-Jun was unaffected. Nifedipine 35-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 175-180 10514457-10 1999 These observations suggest that calcium influx through L-type channels is required for GnRH-induced activation of ERK and c-Fos and that the influence of calcium lies downstream of protein kinase C. Calcium 32-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 10506111-3 1999 In this study, the monoterpene perillyl alcohol (POH) was found to induce transient expression of the c-jun and c-fos genes transcriptionally. Monoterpenes 19-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 10506111-3 1999 In this study, the monoterpene perillyl alcohol (POH) was found to induce transient expression of the c-jun and c-fos genes transcriptionally. perillyl alcohol 31-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 10506111-3 1999 In this study, the monoterpene perillyl alcohol (POH) was found to induce transient expression of the c-jun and c-fos genes transcriptionally. perillyl alcohol 49-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 10512366-8 1999 Finally, both palmitate and oleate caused c-fos and nur-77 mRNA accumulation in isolated rat islets. Palmitates 14-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 10502313-6 1999 In addition to the LC, 8-OH-DPAT (0.1 mg/kg) also induced a marked increase in Fos-LI in various forebrain areas including the medial prefrontal cortex (infralimbic and cingulate cortical areas). 8-Hydroxy-2-(di-n-propylamino)tetralin 23-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 10502313-7 1999 More detailed analysis of the Fos response to 8-OH-DPAT in the medial prefrontal cortex revealed that the effect was attenuated by pretreatment with a combination of the beta(1)- and beta(2)-adrenoceptor antagonists ICI 118551 (4 mg/kg) and metoprolol (4 mg/kg), but not the alpha(1)-adrenoceptor antagonist prazosin (5 mg/kg). 8-Hydroxy-2-(di-n-propylamino)tetralin 46-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 10502313-7 1999 More detailed analysis of the Fos response to 8-OH-DPAT in the medial prefrontal cortex revealed that the effect was attenuated by pretreatment with a combination of the beta(1)- and beta(2)-adrenoceptor antagonists ICI 118551 (4 mg/kg) and metoprolol (4 mg/kg), but not the alpha(1)-adrenoceptor antagonist prazosin (5 mg/kg). Metoprolol 241-251 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 10502313-7 1999 More detailed analysis of the Fos response to 8-OH-DPAT in the medial prefrontal cortex revealed that the effect was attenuated by pretreatment with a combination of the beta(1)- and beta(2)-adrenoceptor antagonists ICI 118551 (4 mg/kg) and metoprolol (4 mg/kg), but not the alpha(1)-adrenoceptor antagonist prazosin (5 mg/kg). Prazosin 308-316 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 10512366-8 1999 Finally, both palmitate and oleate caused c-fos and nur-77 mRNA accumulation in isolated rat islets. Oleic Acid 28-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 10512366-0 1999 Palmitate and oleate induce the immediate-early response genes c-fos and nur-77 in the pancreatic beta-cell line INS-1. Oleic Acid 14-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 10548676-8 1999 Treatment of rats with irbesartan significantly improved neurological outcome of focal cerebral ischemia when compared with the vehicle-treated group and markedly reduced the expression of c-Fos and c-Jun proteins in the cortex on the ligated side of the brain. Irbesartan 23-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 189-194 10548676-9 1999 Irbesartan pretreatment completely abolished the ischemia-induced c-Fos expression in the hippocampus. Irbesartan 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 10512366-2 1999 Palmitate and oleate, but not long-chain polyunsaturated fatty acids, caused a pronounced accumulation of c-fos and nur-77 mRNAs in beta-cells (INS cells) to an extent similar to that produced by the protein kinase C (PKC) activator phorbol myristate acetate (PMA). Palmitates 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 10512366-2 1999 Palmitate and oleate, but not long-chain polyunsaturated fatty acids, caused a pronounced accumulation of c-fos and nur-77 mRNAs in beta-cells (INS cells) to an extent similar to that produced by the protein kinase C (PKC) activator phorbol myristate acetate (PMA). Oleic Acid 14-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 10498647-10 1999 Expression of c-fos in response to PDGF was also reduced, but not suppressed, by treatment with PD98059. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 96-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 10519059-11 1999 These data suggest that Fos stimulus-transcription cascade is activated in these cells by decreased central availability of this monocarboxylate fuel, and that cellular sources of regulatory signaling of lactate utilization exist within the periventricular CNS. monocarboxylate 129-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 10526114-2 1999 To determine if the periaqueductal gray (PAG) and the dorsal raphe nucleus (DRN) are involved in morphine-induced c-Fos and JunB expression in the caudate-putamen (CPu), the mu receptor antagonist, beta-funaltrexamine (beta-FNA), was unilaterally infused into the PAG adjacent to DRN prior to morphine. Morphine 97-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 10526114-4 1999 In the CPu of beta-FNA treated rats, morphine-induced c-Fos and JunB were attenuated compared to vehicle-infused rats. beta-funaltrexamine 14-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 10526114-4 1999 In the CPu of beta-FNA treated rats, morphine-induced c-Fos and JunB were attenuated compared to vehicle-infused rats. Morphine 37-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 10526114-5 1999 These results suggest that morphine acts within the PAG-DRN to exert rapid behavioral effects and to induce c-Fos and JunB in the striatum. Morphine 27-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 10479714-0 1999 The effects of acute nicotine on the metabolism of dopamine and the expression of Fos protein in striatal and limbic brain areas of rats during chronic nicotine infusion and its withdrawal. Nicotine 21-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 10479714-0 1999 The effects of acute nicotine on the metabolism of dopamine and the expression of Fos protein in striatal and limbic brain areas of rats during chronic nicotine infusion and its withdrawal. Nicotine 152-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 10479714-7 1999 Acute nicotine increased Fos immunostaining (IS) in the caudate-putamen (CPU), the core of nucleus accumbens (NAcc), the cingulate cortex (Cg), and the central nucleus of amygdala (ACe) significantly. Nicotine 6-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 10484482-0 1999 Duodenal loading with glucose induces fos expression in rat brain: selective blockade by devazepide. Glucose 22-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 10484482-0 1999 Duodenal loading with glucose induces fos expression in rat brain: selective blockade by devazepide. Devazepide 89-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 10484482-1 1999 The role of CCK in mediating neuronal activity in the brain in response to dietary carbohydrate was measured by detecting Fos immunoreactivity in response to duodenal glucose load in rats after administration of the CCK-A receptor antagonist devazepide. Glucose 167-174 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 10484482-2 1999 In adult, male Sprague-Dawley rats, infusion for 30 min of 545 mg (2.18 kcal) dextrose through a duodenal cannula induced Fos expression in the nucleus of the solitary tract (NTS), area postrema (AP), lateral division of the central nucleus of the amygdala (CeAL), and the external subnucleus of the lateral parabrachial nucleus (LPBE). Glucose 78-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 10513588-0 1999 Environmental modulation of amphetamine-induced c-fos expression in D1 versus D2 striatal neurons. Amphetamine 28-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 10513588-1 1999 We have reported previously that exposure to environmental novelty enhances the behavioral activating effects of amphetamine and its ability to induce the immediate early gene c-fos in the striatum and in other brain regions. Amphetamine 113-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 176-181 10513588-2 1999 In the present study, we used double in situ hybridization histochemistry to study the effect of amphetamine and/or novelty on c-fos expression in two populations of striatal neurons that preferentially express either D1 or D2 dopamine receptor mRNA. Amphetamine 97-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 10513588-3 1999 When given intraperitoneally to rats in their home cage, amphetamine (2.0 mg/kg) increased c-fos expression only in D1 neurons. Amphetamine 57-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 10513588-4 1999 In contrast, when the same dose of amphetamine was administered to rats in a novel environment, c-fos was increased in both D1 and D2 neurons. Amphetamine 35-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 10486183-0 1999 Effects of amphetamine and cocaine treatment on c-Fos, Jun-B, and Krox-24 expression in rats with intrastriatal dopaminergic grafts. Cocaine 27-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 10486183-2 1999 d-Amphetamine-induced c-fos activation is reduced in the neostriatum deprived of DA afferents. Dextroamphetamine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 10486183-3 1999 Dopaminergic grafts implanted into the denervated neostriatum induce a c-fos hyperexpression when challenged with d-amphetamine, which is correlated with the exaggerated compensation of d-amphetamine-induced rotation. Dextroamphetamine 114-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 10486183-3 1999 Dopaminergic grafts implanted into the denervated neostriatum induce a c-fos hyperexpression when challenged with d-amphetamine, which is correlated with the exaggerated compensation of d-amphetamine-induced rotation. Dextroamphetamine 186-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 10486183-7 1999 A c-fos hyperexpression was observed within the grafted neostriatum, which was correlated with the compensation of d-amphetamine- or cocaine-induced rotation. Dextroamphetamine 115-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 2-7 10486183-7 1999 A c-fos hyperexpression was observed within the grafted neostriatum, which was correlated with the compensation of d-amphetamine- or cocaine-induced rotation. Cocaine 133-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 2-7 10486183-13 1999 This generalized c-fos overshoot indicates an abnormal activation of postsynaptic neurons by dopamine and points to its value as an indicator of the deleterious effects of DA grafts. Dopamine 93-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 10486183-13 1999 This generalized c-fos overshoot indicates an abnormal activation of postsynaptic neurons by dopamine and points to its value as an indicator of the deleterious effects of DA grafts. Dopamine 172-174 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 10526780-2 1999 We observed the effects of dietary heme-iron and short chain frucooligosaccharides (Sc-FOS) in relation to prevention of postgastrectomy anemia in rats. frucooligosaccharides 61-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 10526780-8 1999 Dietary Sc-FOS increase iron absorption and thereby prevented completely this anemia in gastrectomized rats fed the control diet but this effect of Sc-FOS in rats fed heme diet was not complete. Iron 24-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 10569236-0 1999 Pharmacological manipulation of central nitric oxide/guanylate cyclase activity alters Fos expression by rat hypothalamic vasopressinergic neurons during acute glucose deprivation. Nitric Oxide 40-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 10569236-4 1999 Adult male rats pretreated by intraventricular administration of saline exhibited extensive colabeling of vasopressinergic neurons in both brain sites for Fos following systemic injection of the glucose antimetabolite, 2-deoxy-D-glucose. Sodium Chloride 65-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-158 10569236-4 1999 Adult male rats pretreated by intraventricular administration of saline exhibited extensive colabeling of vasopressinergic neurons in both brain sites for Fos following systemic injection of the glucose antimetabolite, 2-deoxy-D-glucose. Glucose 195-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-158 10569236-4 1999 Adult male rats pretreated by intraventricular administration of saline exhibited extensive colabeling of vasopressinergic neurons in both brain sites for Fos following systemic injection of the glucose antimetabolite, 2-deoxy-D-glucose. Deoxyglucose 219-236 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-158 10569236-8 1999 coadministration of SIN1 and the nitric-oxide sensitive guanylate cyclase inhibitor, ODQ, prior to the antimetabolite reversed these inhibitory effects of SIN1 on Fos expression by these cells. nitric- 33-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 10569236-8 1999 coadministration of SIN1 and the nitric-oxide sensitive guanylate cyclase inhibitor, ODQ, prior to the antimetabolite reversed these inhibitory effects of SIN1 on Fos expression by these cells. 1H-(1,2,3)oxadiazolo(4,4-a)quinoxalin-1-one 85-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 10569236-10 1999 The present studies demonstrate that exogenous activation of the nitric oxide/guanylate cyclase/cGMP pathway in the brain inhibits nuclear accumulation of the AP-1 transcription factor, Fos, in vasopressinergic neurons during cellular glucopenia, and suggest that this neurotransmitter is critical for transactivational effects of glucoprivation on these neuropeptidergic neurons. Nitric Oxide 65-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 186-189 10569236-10 1999 The present studies demonstrate that exogenous activation of the nitric oxide/guanylate cyclase/cGMP pathway in the brain inhibits nuclear accumulation of the AP-1 transcription factor, Fos, in vasopressinergic neurons during cellular glucopenia, and suggest that this neurotransmitter is critical for transactivational effects of glucoprivation on these neuropeptidergic neurons. Cyclic GMP 96-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 186-189 10462695-0 1999 MK-801-induced expression of Fos protein family members in the rat retrosplenial granular cortex. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 10462695-1 1999 The N-methyl-D-aspartate (NMDA) receptor antagonist MK-801 has been shown to induce an acute Fos and Fos-related antigen (Fra) expression in the rat retrosplenial granular cortex (RSG), but the exact composition of the Fos protein family and their individual dynamic alterations are unknown. Dizocilpine Maleate 52-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 10462695-1 1999 The N-methyl-D-aspartate (NMDA) receptor antagonist MK-801 has been shown to induce an acute Fos and Fos-related antigen (Fra) expression in the rat retrosplenial granular cortex (RSG), but the exact composition of the Fos protein family and their individual dynamic alterations are unknown. Dizocilpine Maleate 52-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-104 10513171-0 1999 [The study of the anesthetic action of halothane on the rat spinal cord by fos immunoreactivity]. Halothane 39-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 10513171-1 1999 This study was performed to examine the anesthetic action of halothane on the spinal cord of rats by using fos immunoreactivity, a marker for neuronal activity following noxious stimulation. Halothane 61-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 10513171-9 1999 The number of fos immunoreactive cells decreased in the cord of rats that showed no response to noxious stimulation by halothane 1 or 1.5 MAC. Halothane 119-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 10471092-0 1999 Pseudoephedrine, a sympathomimetic agent, induces Fos-like immunoreactivity in rat nucleus accumbens and striatum. Pseudoephedrine 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 10471092-2 1999 Following intraperitoneal injection of Sprague Dawley rats with pseudoephedrine, Fos induction was measured in various brain areas by Western blots and immunocytochemistry. Pseudoephedrine 64-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-84 10471092-3 1999 Pseudoephedrine induced Fos-like immunoreactivity in the nucleus accumbens and striatum in a time and concentration-dependent manner with maximal effect at 60 mg/kg 2 h after injection. Pseudoephedrine 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 10471092-5 1999 Pre-injection with the D1 dopamine receptor antagonist SCH23390 partially and completely blocked pseudoephedrine-induced Fos-like immunoreactivity in the striatum and nucleus accumbens, respectively, suggesting that the action of pseudoephedrine is mediated via dopamine release and results in the activation of D1 dopamine receptors. SCH 23390 55-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-124 10471092-5 1999 Pre-injection with the D1 dopamine receptor antagonist SCH23390 partially and completely blocked pseudoephedrine-induced Fos-like immunoreactivity in the striatum and nucleus accumbens, respectively, suggesting that the action of pseudoephedrine is mediated via dopamine release and results in the activation of D1 dopamine receptors. Pseudoephedrine 97-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-124 10471092-5 1999 Pre-injection with the D1 dopamine receptor antagonist SCH23390 partially and completely blocked pseudoephedrine-induced Fos-like immunoreactivity in the striatum and nucleus accumbens, respectively, suggesting that the action of pseudoephedrine is mediated via dopamine release and results in the activation of D1 dopamine receptors. Dopamine 26-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-124 10491822-0 1999 Intrathecal treatment with MK-801 suppresses thermal nociceptive responses and prevents c-fos immunoreactivity induced in rat lumbar spinal cord neurons. Dizocilpine Maleate 27-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 10491822-4 1999 In addition, the influence of MK-801 on c-Fos immunoreactivity in the rat lumbar spinal cord neurons after the peripheral noxious heat was examined. Dizocilpine Maleate 30-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 10491822-8 1999 MK-801 significantly increased the thermal withdrawal threshold by 60% following noxious heat stimulation and reduced c-Fos immunoreactivity in the second order neurons by 70% in the dorsal horn. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 10491822-9 1999 The study suggests that glutamate plays a pivotal role in the thermal nociceptive pathway and indicates that the NMDA receptor is necessary to maintain normal thermal sensitization, possibly by regulating c-fos gene expression in second order neurons. Glutamic Acid 24-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 205-210 11498949-4 1999 The angiotensin II-induced expression of c-fos gene was also blocked by Ca2+ channel antagonist nicardipine. Nicardipine 96-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 10580296-0 1999 c-Fos expression in the myenteric plexus, spinal cord and brainstem following injection of formalin in the rat colonic wall. Formaldehyde 91-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 10580296-1 1999 Fos expression induced by injection of dilute formalin (50 microl, 5% in physiological saline) into the colonic wall was examined in the myenteric plexus, lumbosacral spinal cord and brainstem of the rat. Formaldehyde 46-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 10580296-1 1999 Fos expression induced by injection of dilute formalin (50 microl, 5% in physiological saline) into the colonic wall was examined in the myenteric plexus, lumbosacral spinal cord and brainstem of the rat. Sodium Chloride 87-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 10580296-2 1999 The aims of this study were (i) to determine whether neurons in these regions express Fos in response to the injection of formalin into the colon and (ii) to examine whether administration of an alpha 2 adrenoceptor agonist modulates Fos expression. Formaldehyde 122-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 10580296-4 1999 Saline injected in the colon induced Fos in enteric glia in the myenteric plexus. Sodium Chloride 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 10580296-5 1999 The number of Fos immunoreactive nuclei significantly increased in both myenteric neurons and enteric glia after the injection of formalin. Formaldehyde 130-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 10580296-6 1999 Similarly, Fos immunoreactive neuronal nuclei were significantly increased in the spinal cord, area postrema and nucleus of the solitary tract after the injection of formalin. Formaldehyde 166-174 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 10580296-7 1999 Pretreatment of rats with the alpha 2 adrenoceptor agonist xylazine (2, 4 and 8 mg/kg) 15 min before the injection of formalin, dose-dependently reduced the number of Fos immunoreactive neuronal and glial nuclei in the myenteric plexus, and neuronal nuclei in the spinal cord and brainstem. Xylazine 59-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 167-170 10580296-9 1999 These data show that injection of formalin in the colonic wall results in Fos expression in myenteric neurons and enteric glia, and neurons in the spinal cord and brainstem. Formaldehyde 34-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 11130958-0 1999 c-Fos expression in the myenteric plexus, spinal cord and brainstem following injection of formalin in the rat colonic wall. Formaldehyde 91-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 11130958-1 1999 Fos expression induced by injection of dilute formalin (50 microl, 5% in physiological saline) into the colonic wall was examined in the myenteric plexus, lumbosacral spinal cord and brainstem of the rat. Formaldehyde 46-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 11130958-1 1999 Fos expression induced by injection of dilute formalin (50 microl, 5% in physiological saline) into the colonic wall was examined in the myenteric plexus, lumbosacral spinal cord and brainstem of the rat. Sodium Chloride 87-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 11130958-2 1999 The aims of this study were (i) to determine whether neurons in these regions express Fos in response to the injection of formalin into the colon and (ii) to examine whether administration of an alpha 2 adrenoceptor agonist modulates Fos expression. Formaldehyde 122-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 11130958-4 1999 Saline injected in the colon induced Fos in enteric glia in the myenteric plexus. Sodium Chloride 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 11130958-5 1999 The number of Fos immunoreactive nuclei significantly increased in both myenteric neurons and enteric glia after the injection of formalin. Formaldehyde 130-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 11130958-6 1999 Similarly, Fos immunoreactive neuronal nuclei were significantly increased in the spinal cord, area postrema and nucleus of the solitary tract after the injection of formalin. Formaldehyde 166-174 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 11130958-7 1999 Pretreatment of rats with the alpha 2 adrenoceptor agonist xylazine (2, 4 and 8 mg/kg) 15 min before the injection of formalin, dose-dependently reduced the number of Fos immunoreactive neuronal and glial nuclei in the myenteric plexus, and neuronal nuclei in the spinal cord and brainstem. Xylazine 59-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 167-170 11130958-9 1999 These data show that injection of formalin in the colonic wall results in Fos expression in myenteric neurons and enteric glia, and neurons in the spinal cord and brainstem. Formaldehyde 34-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 10521594-0 1999 Long-lasting sensitization towards morphine in motoric and limbic areas as determined by c-fos expression in rat brain. Morphine 35-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 10521594-4 1999 To identify brain areas involved in these long-lasting processes, we studied the expression of the transcription factor c-fos by in situ hybridization in rat brain as a marker for changes in gene expression after single or repeated morphine applications in the animals. Morphine 232-240 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 10521594-5 1999 The only c-fos signal that exceeded background after a single dose of morphine (50 mg/kg) was a diffuse expression in the lateral septum. Morphine 70-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 10521594-6 1999 In contrast, repeated dosage twice daily for 10 days and ascending from 10 to 50 mg/kg resulted in a sharply delineated morphine-induced c-fos synthesis in the dorsomedial and lateral striatum, lateral septum, medial mammillary nuclei, anterior thalamus and, in part masked by a high background due to injection stress, in the cingulate cortex. Morphine 120-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 10521594-8 1999 The c-fos response was inducible by morphine in pretreated animals for up to 8 weeks after finishing the repeated application scheme. Morphine 36-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 10521594-10 1999 Therefore, the sensitization of morphine-induced c-fos expression in parts of the striatum seems to correlate with the locomotor effects of repeated morphine application, whereas the observed sensitization in several limbic brain areas might reflect emotional phenomena like increased self-administration in rats or drug craving in humans. Morphine 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 10521594-10 1999 Therefore, the sensitization of morphine-induced c-fos expression in parts of the striatum seems to correlate with the locomotor effects of repeated morphine application, whereas the observed sensitization in several limbic brain areas might reflect emotional phenomena like increased self-administration in rats or drug craving in humans. Morphine 149-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 10484482-3 1999 Devazepide treatment (1 mg/kg) attenuated Fos expression in the NTS and AP by 81 and 78%, respectively, but not in the CeAL or LPBE. Devazepide 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 10497900-0 1999 Opioidergic and adrenergic modulation of formalin-evoked spinal c-fos mRNA expression and nocifensive behavior in the rat. Formaldehyde 41-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 10497900-2 1999 We determine whether formalin-induced spinal c-fos mRNA expression (Northern blotting) correlates with nocifensive behavior following pretreatment with morphine, the alpha2-adrenoceptor agonist dexmedetomidine, or their respective antagonists naloxone and atipamezole. Formaldehyde 21-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 10497900-2 1999 We determine whether formalin-induced spinal c-fos mRNA expression (Northern blotting) correlates with nocifensive behavior following pretreatment with morphine, the alpha2-adrenoceptor agonist dexmedetomidine, or their respective antagonists naloxone and atipamezole. Morphine 152-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 10497900-3 1999 Both opiate and alpha2-adrenoceptor agonists reduced formalin-induced c-fos gene transcription and nocifensive behavior via their cognate receptors. Formaldehyde 53-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 10497900-4 1999 Unexpectedly, blockade of either the opiate or alpha2-adrenergic receptors, alone, caused an increase in formalin-evoked c-fos mRNA; while blocking the opiate receptor had no effect on formalin-induced behavior, alpha2-adrenoceptor block had an analgesic effect, indicating discordance between c-fos message transcription and nocifensive behavior. Formaldehyde 105-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-126 10497900-4 1999 Unexpectedly, blockade of either the opiate or alpha2-adrenergic receptors, alone, caused an increase in formalin-evoked c-fos mRNA; while blocking the opiate receptor had no effect on formalin-induced behavior, alpha2-adrenoceptor block had an analgesic effect, indicating discordance between c-fos message transcription and nocifensive behavior. Formaldehyde 105-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 294-299 10497900-5 1999 We concluded that the formalin-induced spinal c-fos signal was a poor predictor of the behavioral response to pharmacological manipulation of pain processing pathways. Formaldehyde 22-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 10482817-0 1999 Fos expression in gonadotropin-releasing hormone neurons by naloxone or bicuculline in intact male rats. Naloxone 60-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 10482794-0 1999 Transient MRI-detected water apparent diffusion coefficient reduction correlates with c-fos mRNA but not hsp70 mRNA induction during focal cerebral ischemia in rats. Water 23-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 10482794-6 1999 The most significant finding of this work was that for both durations of ischemia, c-fos induction was observed in cortical and sub-cortical regions exhibiting a transient reduction in the apparent diffusion coefficient of water (ADC). Water 223-228 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 10505476-0 1999 CO2-induced c-fos expression in medullary neurons during early development. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 10505476-5 1999 No age related differences were observed in the number of neurons exhibiting CO2-induced Fos expression. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 77-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 10446216-2 1999 Inhibition of PI 3-kinase by LY294002 or wortmannin, two specific PI 3-kinase antagonists, or co-transfection with a dominant negative mutant of PI 3-kinase dose-dependently blocked stimulation of c-fos SRE by TNF-alpha. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 29-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 197-202 10446216-2 1999 Inhibition of PI 3-kinase by LY294002 or wortmannin, two specific PI 3-kinase antagonists, or co-transfection with a dominant negative mutant of PI 3-kinase dose-dependently blocked stimulation of c-fos SRE by TNF-alpha. Wortmannin 41-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 197-202 10404113-1 1999 We used immunocytochemistry to determine the regional and temporal distribution of Fos protein expression in awake and unrestrained rats after a unilateral stereotaxic microinjection of a cholinergic agonist, carbachol, in the thalamic ventroposterolateral and reticular nuclei, previously shown to cause limbic and generalized convulsive seizures. Carbachol 209-218 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 10404113-2 1999 The microinjection of carbachol elicits behavioral alterations including immobilization, staring, facial and jaw clonus, rearing, and falling, followed by recurrent generalized convulsive seizures, and a pattern of c-fos expression throughout the brain. Carbachol 22-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 215-220 10521585-0 1999 Acute injection of drugs with low addictive potential (delta(9)-tetrahydrocannabinol, 3,4-methylenedioxymethamphetamine, lysergic acid diamide) causes a much higher c-fos expression in limbic brain areas than highly addicting drugs (cocaine and morphine). Dronabinol 55-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 10521585-0 1999 Acute injection of drugs with low addictive potential (delta(9)-tetrahydrocannabinol, 3,4-methylenedioxymethamphetamine, lysergic acid diamide) causes a much higher c-fos expression in limbic brain areas than highly addicting drugs (cocaine and morphine). N-Methyl-3,4-methylenedioxyamphetamine 86-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 10482817-0 1999 Fos expression in gonadotropin-releasing hormone neurons by naloxone or bicuculline in intact male rats. Bicuculline 72-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 10482817-3 1999 After the naloxone infusion, some Fos-positive GnRH neurons were found from the preoptic area to the basal hypothalamic area, whereas those were found from the diagonal band of Broca to the preoptic area after the bicuculline infusion. Naloxone 10-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 10446338-0 1999 Infusion of beta-FNA into the thalamus attenuates morphine-induced c-Fos induction in the rat caudate putamen. Morphine 50-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 10521585-0 1999 Acute injection of drugs with low addictive potential (delta(9)-tetrahydrocannabinol, 3,4-methylenedioxymethamphetamine, lysergic acid diamide) causes a much higher c-fos expression in limbic brain areas than highly addicting drugs (cocaine and morphine). lysergic acid diamide 121-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 10521585-0 1999 Acute injection of drugs with low addictive potential (delta(9)-tetrahydrocannabinol, 3,4-methylenedioxymethamphetamine, lysergic acid diamide) causes a much higher c-fos expression in limbic brain areas than highly addicting drugs (cocaine and morphine). Cocaine 233-240 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 10521585-0 1999 Acute injection of drugs with low addictive potential (delta(9)-tetrahydrocannabinol, 3,4-methylenedioxymethamphetamine, lysergic acid diamide) causes a much higher c-fos expression in limbic brain areas than highly addicting drugs (cocaine and morphine). Morphine 245-253 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 10521585-4 1999 Cocaine in a high dose of 50 mg/kg yielded only a discrete c-fos expression in the medial and central striatum. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 10521585-5 1999 Morphine (50 mg/kg) caused a weak c-fos synthesis in the lateral septum. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 10521585-6 1999 THC (delta(9)-tetrahydrocannabinol), 25 mg/kg, induced c-fos mRNA again in the lateral septum and furthermore in large parts of the striatum including the nucleus accumbens. Dronabinol 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 10521585-6 1999 THC (delta(9)-tetrahydrocannabinol), 25 mg/kg, induced c-fos mRNA again in the lateral septum and furthermore in large parts of the striatum including the nucleus accumbens. Dronabinol 5-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 10521585-7 1999 LSD (lysergic acid diamide), 1 mg/kg, elicited a similar c-fos expression pattern as THC, but there was additionally a very strong hybridization signal in the cerebral cortex, especially in the upper layers, and in the ventral part of the periaqueductal gray. lysergic acid diamide 5-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 10521585-10 1999 Taken together, our study revealed that the drugs with the highest addictive potential, cocaine and morphine, yielded a very low c-fos synthesis throughout the brain whereas the brain regions closely linked to pleasure (especially the nucleus accumbens) responded strongly to drugs with an apparently lower addictive potential (THC, LSD, MDMA). Cocaine 88-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 10521585-10 1999 Taken together, our study revealed that the drugs with the highest addictive potential, cocaine and morphine, yielded a very low c-fos synthesis throughout the brain whereas the brain regions closely linked to pleasure (especially the nucleus accumbens) responded strongly to drugs with an apparently lower addictive potential (THC, LSD, MDMA). Morphine 100-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 10446338-0 1999 Infusion of beta-FNA into the thalamus attenuates morphine-induced c-Fos induction in the rat caudate putamen. beta-funaltrexamine 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 10446338-2 1999 Morphine-induced c-Fos expression in the CPu has been shown to be blocked by pretreatment with antagonists to N-methyl-D-aspartate receptors, indicating the involvement of glutamate in this morphine-induced response. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 10446338-2 1999 Morphine-induced c-Fos expression in the CPu has been shown to be blocked by pretreatment with antagonists to N-methyl-D-aspartate receptors, indicating the involvement of glutamate in this morphine-induced response. Glutamic Acid 172-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 10446338-2 1999 Morphine-induced c-Fos expression in the CPu has been shown to be blocked by pretreatment with antagonists to N-methyl-D-aspartate receptors, indicating the involvement of glutamate in this morphine-induced response. Morphine 190-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 10446338-4 1999 Infusion of beta-FNA near specific medial thalamic nuclei attenuated morphine-induced c-Fos expression in the CPu. Morphine 69-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 10444561-0 1999 Fos expression in brain stem nuclei of pregnant rats after hydralazine-induced hypotension. Hydralazine 59-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 10444567-7 1999 The majority of GLP-1 neurons were activated to express c-Fos after LiCl, LPS, or CCK treatment, including (in LiCl-treated rats) those projecting to the PVN. Lithium Chloride 68-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 10430646-8 1999 In vitro, ROS 17/2.8 cells expressed detectable levels of c-fos, c-jun, c-myc, OC, OP, ALP, COL1A1, and PTHR but not MMP-9. ros 10-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 10428063-5 1999 As extracellular signal-regulated kinase (Erk) and stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) phosphorylation may induce Fos and Jun gene transcription and activator protein-1 (AP-1) DNA binding, the activation of Erk1, Erk2, p38, and SAPK/JNK was examined in the nucleus tractus solitarii and neocortex during hypoxia and following administration of MK-801. Dizocilpine Maleate 375-381 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-148 10404113-3 1999 In addition to the hypothalamic paraventricular and supraoptic nuclei, the initial induction of c-fos expression was observed as early as 15 minutes after the carbachol microinjection, in the piriform and entorhinal cortices, the thalamic paraventricular, the supramammilary, the lateral parabrachial nuclei, and the central gray. Carbachol 159-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 10452460-5 1999 Amphetamine-induced production of the immediate early gene protein product Fos was quantified to determine neuronal dopaminergic response in caudate-putamen (striatum). Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 10452460-8 1999 Five days after injury, ipsilateral striatal Fos expression was reduced by 51% in TBI rats experiencing normal light cycling (p < 0.006). tbi 82-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 10419543-4 1999 Injections of DA produce a strong and prolonged activated protein 1 (AP-1) activity that contains c-fos, c-jun, and phosphorylated c-jun protein. Dopamine 14-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 10461024-5 1999 Prior administration of morphine or GHRH significantly reduced the number of Fos-positive cells in the arcuate nucleus of rats injected with either GHS, although when given together, morphine and GHRH did not produce a greater reduction in Fos expression than when given alone. Morphine 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-80 10467924-11 1999 injection of SR141716A increased Fos expression in both normal and inflamed animals, to a different extent in different laminae. Rimonabant 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 10467924-12 1999 In normal animals, the increase was primarily in laminae V-VI and in the ventral horn; in animals with persistent inflammation SR141716A increased the number of Fos neurons in laminae I-II and V-VI. Rimonabant 127-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-164 10462181-0 1999 Effects of perospirone, a novel 5-HT2 and D2 receptor antagonist, on Fos protein expression in the rat forebrain. perospirone 11-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 10467924-13 1999 These results demonstrate that WIN55212-2 reverses inflammation-induced allodynia at doses that do not produce analgesia and that SR141716A differentially affects the pattern of Fos expression in the spinal cord, depending on the presence or absence of inflammation. Rimonabant 130-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 178-181 10462181-1 1999 The effects of perospirone, a novel 5-HT2 and D2 receptor antagonist, on Fos protein expression in the nucleus accumbens (NA) and dorsolateral striatum (DLSt) were compared with those of typical (i.e., haloperidol and fluphenazine) and atypical (i.e., clozapine and risperidone) antipsychotics using immunohistochemical techniques in rats. perospirone 15-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-76 10455919-0 1999 Antisense oligonucleotides to c-fos and c-jun inhibit intimal thickening in a rat vein graft model. Oligonucleotides 10-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 10462181-2 1999 Perospirone and other antipsychotics tested at doses that exerted D2 blocking actions increased Fos-like immunoreactivity both in the NA and DLSt. perospirone 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-99 10462181-3 1999 However, the levels of Fos expression in the DLSt induced by perospirone and clozapine were less than those induced by haloperidol and fluphenazine. perospirone 61-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 10462181-3 1999 However, the levels of Fos expression in the DLSt induced by perospirone and clozapine were less than those induced by haloperidol and fluphenazine. Clozapine 77-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 10462181-4 1999 When compared the differences in numbers of Fos-positive neurons between in the NA and DLSt, perospirone, clozapine, and risperidone preferentially increased Fos expression in the NA. perospirone 93-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 10462181-4 1999 When compared the differences in numbers of Fos-positive neurons between in the NA and DLSt, perospirone, clozapine, and risperidone preferentially increased Fos expression in the NA. perospirone 93-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-161 10462181-4 1999 When compared the differences in numbers of Fos-positive neurons between in the NA and DLSt, perospirone, clozapine, and risperidone preferentially increased Fos expression in the NA. Clozapine 106-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 10462181-4 1999 When compared the differences in numbers of Fos-positive neurons between in the NA and DLSt, perospirone, clozapine, and risperidone preferentially increased Fos expression in the NA. Clozapine 106-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-161 10462181-4 1999 When compared the differences in numbers of Fos-positive neurons between in the NA and DLSt, perospirone, clozapine, and risperidone preferentially increased Fos expression in the NA. Risperidone 121-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 10462181-4 1999 When compared the differences in numbers of Fos-positive neurons between in the NA and DLSt, perospirone, clozapine, and risperidone preferentially increased Fos expression in the NA. Risperidone 121-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-161 10462181-5 1999 These findings suggest that perospirone has a preferential action on the mesolimbic (vs. nigrostriatal) dopaminergic system in inducing Fos protein in the rat brain, which may be related to its atypical antipsychotic properties. perospirone 28-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-139 10455919-5 1999 Additional rats underwent bypasses and at the time of the procedure 1 graft was treated with a pluronic gel containing an ASO to c-fos, c-jun, or sense and the contralateral side was treated with pluronic gel only. Poloxamer 95-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 10409647-4 1999 Although both depolarization-induced calcium influx and bradykinin stimulation of PC12 cells were found to induce c-fos transcription through EGFR activation, the former signal is CaM kinase-dependent and the latter was shown to be independent. Calcium 37-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 10678109-3 1999 RESULTS: The increases of NOS-positive neurons, Fos, NOS/Fos double labelling neurons of the spinal dorsal horn and the PIR after formalin injection were markedly inhibited by intrathecal injecting (ith) Cor (0.5-1.5 U), which were obviously attenuated by L-arginine (Arg, 5-15 nmol, ith), the substrate of NOS. Formaldehyde 130-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 10478560-3 1999 Desensitization was assessed by the decrease in the number of Fos-immunoreactive spinal neurons induced by the intravesical instillation of 1% acetic acid, when the latter was preceded by resiniferatoxin in concentrations between 1 and 1000 nM. Acetic Acid 143-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 10478560-4 1999 Irritation, as shown by the noxious excitation of vesical sensory innervation, was measured by the c-fos response evoked by a single application of resiniferatoxin. resiniferatoxin 148-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 10478560-5 1999 As to the desensitizing power, resiniferatoxin produced a dose-dependent effect with a maximum at 100 nM, which decreased Fos-immunoreactive cell numbers to less than 10% of controls. resiniferatoxin 31-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 10478560-7 1999 As to the irritating power, the saturation dose of resiniferatoxin (100 nM) produced a very weak c-fos activation in lumbosacral spinal cord segments. resiniferatoxin 51-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 10415375-1 1999 Previous studies from this laboratory have demonstrated that acute, systemic administration of morphine results in an induction of the immediate-early gene (IEG) proteins, c-Fos and Jun-B, in the dorsomedial portion of the rat caudate-putamen (CPu). Morphine 95-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-177 10415375-2 1999 These studies have also shown that morphine can induce c-Fos in the central medial nucleus of the thalamus (CM). Morphine 35-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 10415375-5 1999 As compared to an acute dose of morphine in a naive animal, the induction of c-Fos was increased in the dorsolateral CPu following challenge injection at 7 days, but not at 14 days. Morphine 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 10454158-0 1999 Ketamine suppresses c-fos expression in dorsal horn neurons after acute constrictive sciatic nerve injury in the rat. Ketamine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 10454158-3 1999 Fos immunoreactive neurons have a unique response pattern to neuropathic pain which is sensitive to ketamine. Ketamine 100-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 10657505-0 1999 Endomorphin-1 reduces carrageenan-induced fos expression in the rat spinal dorsal horn. Carrageenan 22-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 10657505-1 1999 Intraplantar injection of carrageenan induced significant Fos expression in the superficial and deep spinal dorsal horn at the L(4)-L(5)segments and extensive peripheral edema of the ipsilateral foot in rats. Carrageenan 26-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 10657505-2 1999 Intraplantar injection of endomorphin-1, endogenous ligand for mu opioid receptor, in the same region produced dose-dependent reduction of carrageenan-induced Fos expression and peripheral edema, which were completely blocked by co-administration of intraplantar injection of naloxone (20 microgram). Carrageenan 139-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-162 10417396-5 1999 A low concentration (1 micromol/L) of DDC stimulated myocyte growth, as demonstrated by increases in protein synthesis, cellular protein, prepro-atrial natriuretic peptide, and c-fos mRNAs and decreased sarcoplasmic reticulum Ca(2+)ATPase mRNA. Ditiocarb 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 177-182 10397398-6 1999 The 6-OHDAamp and ASF groups exhibited robust, ipsiversive circling behavior, with similar changes in Fos-LI in the striatum, entopeduncular nucleus, superior colliculus, and ventromedial thalamus. 6-ohdaamp 4-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 10391952-4 1999 N-Acetylcysteine also significantly reduced serum-stimulated elevation of c-Fos but did not prevent the normal mitogen-induced increase in c-fos mRNA. Acetylcysteine 0-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 10391952-7 1999 Serum-induced ferricytochrome c reduction, cellular proliferation, and c-Fos elevation were decreased by the flavoprotein-dependent enzyme inhibitor dipheyleneiodonium. dipheyleneiodonium 149-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 10395942-0 1999 Retinoic acid repressed the expression of c-fos and c-jun and induced apoptosis in regenerating rat liver after partial hepatectomy. Tretinoin 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 10395942-4 1999 Northern blot analysis revealed that RA repressed the expression of c-fos and c-jun at 15 and 30 min with the up-regulation of retinoic acid receptor gamma (RARgamma) and RARbeta at 2 h after PH. Tretinoin 37-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 10395942-8 1999 These results suggest that RA exerts the antiproliferative activity only on the early stage of liver regeneration accompanied by the repression of c-fos and c-jun expression and induction of apoptosis. Tretinoin 27-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-152 10383614-3 1999 Exposure to water or removal of the vomeronasal organ suppressed the expression of Fos-ir cells. Water 12-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 10407093-5 1999 A substantial percentage (20%) of dye labeled PGN exhibited Fos-IR after LUT irritation; and a larger percentage (36%) exhibited Fos-IR after electrical stimulation of the pelvic nerve which contains afferent pathways from all of the pelvic organs. pgn 46-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 10407093-7 1999 A selective distribution of different types of neurons was detected in this region: PGN were located ventral to the spinal projection neurons which in turn were located ventral to the majority of unidentified Fos-positive neurons. pgn 84-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 209-212 10407093-8 1999 The distribution of Fos-positive PGN and projection neurons was similar in spinal intact and spinal transected animals indicating that c-fos expression was mediated by monosynaptic afferent input or input from segmental interneurons and was not due to activation of supraspinal micturition reflex pathways. pgn 33-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 10407110-3 1999 The NADPH-d positive cells showed perikarya and cytoplasmic processes laying next to or more frequently apposed to Fos-positive neurons. NADP 4-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-118 10440105-2 1999 Here, we report that the EGF receptor kinase inhibitor AG1478 and the ERK kinase inhibitor PD98059 markedly inhibited angiotensin II-induced c-Fos expression and protein synthesis but not c-Jun expression in these cells. RTKI cpd 55-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 10440105-2 1999 Here, we report that the EGF receptor kinase inhibitor AG1478 and the ERK kinase inhibitor PD98059 markedly inhibited angiotensin II-induced c-Fos expression and protein synthesis but not c-Jun expression in these cells. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 91-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 10409218-4 1999 Atropine and hexamethonium partially decreased c-fos expression (banding vs. banding + atropine/hexamethonium: 700 +/- 67% vs. 400 +/- 67%, P < 0.05). Atropine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 10409218-4 1999 Atropine and hexamethonium partially decreased c-fos expression (banding vs. banding + atropine/hexamethonium: 700 +/- 67% vs. 400 +/- 67%, P < 0.05). Hexamethonium 13-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 10409218-4 1999 Atropine and hexamethonium partially decreased c-fos expression (banding vs. banding + atropine/hexamethonium: 700 +/- 67% vs. 400 +/- 67%, P < 0.05). Atropine 87-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 10409218-4 1999 Atropine and hexamethonium partially decreased c-fos expression (banding vs. banding + atropine/hexamethonium: 700 +/- 67% vs. 400 +/- 67%, P < 0.05). Hexamethonium 96-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 10406836-11 1999 The pressure-induced expression of c-fos was not inhibited by nitrendipine (10 micromol/L), a calcium-free Krebs" solution containing EGTA (1 to 2 mmol/L), calphostin C (0.1 micromol/L), or cytochalasin D (0.4 micromol/L) but was inhibited by genistein (30 micromol/L). Egtazic Acid 134-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 10406835-7 1999 Inhibition of Ang II-induced AP-1 activation with c-fos antisense oligodeoxynucleotide led to a significant reduction of TGF-beta(1) mRNA in VSMCs. Oligodeoxyribonucleotides 66-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 10406836-0 1999 Genistein inhibits pressure-induced expression of c-fos in isolated mesenteric arteries. Genistein 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 10406836-10 1999 Sections of paraffin-embedded arteries were fixed on slides, and the expression of c-fos was determined by in situ hybridization with the use of (35)S-labeled riboprobes. Paraffin 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 10406836-11 1999 The pressure-induced expression of c-fos was not inhibited by nitrendipine (10 micromol/L), a calcium-free Krebs" solution containing EGTA (1 to 2 mmol/L), calphostin C (0.1 micromol/L), or cytochalasin D (0.4 micromol/L) but was inhibited by genistein (30 micromol/L). calphostin C 156-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 10406836-11 1999 The pressure-induced expression of c-fos was not inhibited by nitrendipine (10 micromol/L), a calcium-free Krebs" solution containing EGTA (1 to 2 mmol/L), calphostin C (0.1 micromol/L), or cytochalasin D (0.4 micromol/L) but was inhibited by genistein (30 micromol/L). Cytochalasin D 190-204 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 10406836-11 1999 The pressure-induced expression of c-fos was not inhibited by nitrendipine (10 micromol/L), a calcium-free Krebs" solution containing EGTA (1 to 2 mmol/L), calphostin C (0.1 micromol/L), or cytochalasin D (0.4 micromol/L) but was inhibited by genistein (30 micromol/L). Genistein 243-252 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 10444310-5 1999 Within 30 min, formalin elicited a four- to sixfold increase in plasma ACTH and corticosterone concentrations and intense Fos-like activity was seen in the superficial zones of the lumbar spinal cord ipsilateral to the side of the formalin injection. Formaldehyde 15-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 10444310-5 1999 Within 30 min, formalin elicited a four- to sixfold increase in plasma ACTH and corticosterone concentrations and intense Fos-like activity was seen in the superficial zones of the lumbar spinal cord ipsilateral to the side of the formalin injection. Formaldehyde 231-239 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 10444310-6 1999 In brainstem catecholaminergic neurones, the PVN, and midline thalamic nuclei, formalin-induced Fos-immunopositivity was equally present in the ipsi- and contralateral sides of the injection. Formaldehyde 79-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-99 10412747-5 1999 Lovastatin reduced membrane-bound p21ras and fetal calf serum-induced c-fos and c-jun protein expression. Lovastatin 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 10422655-0 1999 Differential effects of low dose CP122,288 and eletriptan on fos expression due to stimulation of the superior sagittal sinus in cat. cp122 33-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 10422655-0 1999 Differential effects of low dose CP122,288 and eletriptan on fos expression due to stimulation of the superior sagittal sinus in cat. eletriptan 47-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 10485826-6 1999 Only slight expression of c-myc or c-fos was apparent after 30-min oral administration or 1-, 3-, and 6-hr oral administration of catechol. catechol 130-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 10422655-11 1999 In comparison, the clinically effective 5HT(1B/1D) agonist, eletriptan, reduced Fos expression in the trigeminocervical complex to a median of 24 (21-33). eletriptan 60-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 10381370-2 1999 Exposure to crude urine and ultrafiltrated urine preparation (<5000 Da) induced significant Fos expression, which is correlated with cellular activity, in the mitral/tufted cell layer of the accessory olfactory bulb (AOB), while exposure to the remaining substances after ultrafiltration (>5000 Da) and control salt solution did not. Salts 317-321 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 10332800-0 1999 NMDA receptor blockade attenuates the haloperidol induction of Fos protein in the dorsal but not the ventral striatum. Haloperidol 38-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 10406025-0 1999 Antisense oligonucleotides to C-fos reduce postictal seizure susceptibility following fully kindled seizures in rats. Oligonucleotides 10-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 10406025-1 1999 In a previous study we demonstrated that increased FOS expression in the amygdala induced by partial kindling seizures could be attenuated by administering c-Fos specific antisense oligonucleotides. Oligonucleotides 181-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 10406025-1 1999 In a previous study we demonstrated that increased FOS expression in the amygdala induced by partial kindling seizures could be attenuated by administering c-Fos specific antisense oligonucleotides. Oligonucleotides 181-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 10406025-2 1999 In addition, we found that the administration of c-Fos antisense oligonucleotides at the beginning of the amygdala kindling process facilitated the appearance of stage V kindled seizures. Oligonucleotides 65-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 10406025-5 1999 However, c-Fos antisense oligonucleotides significantly reduced the susceptibility to additional ictal events during the postictal refractory period. Oligonucleotides 25-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 10375656-0 1999 Suppression of post-ischemic-induced fos protein expression by an antisense oligonucleotide to c-fos mRNA leads to increased tissue damage. Oligonucleotides 76-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 10375656-0 1999 Suppression of post-ischemic-induced fos protein expression by an antisense oligonucleotide to c-fos mRNA leads to increased tissue damage. Oligonucleotides 76-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 10375656-3 1999 To study the downstream effects of c-fos expression following ischemia, we suppressed the translation of c-fos by administering an antisense oligonucleotide (AO) to c-fos mRNA. Oligonucleotides 141-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 10375656-3 1999 To study the downstream effects of c-fos expression following ischemia, we suppressed the translation of c-fos by administering an antisense oligonucleotide (AO) to c-fos mRNA. Oligonucleotides 141-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 10375656-9 1999 AO administration reduced the number of cells with fci-induced Fos expression by approximately 75%. Oligonucleotides, Antisense 0-2 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 10375656-9 1999 AO administration reduced the number of cells with fci-induced Fos expression by approximately 75%. fci 51-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 10332800-3 1999 In the present study, we examine the role of these receptors in the haloperidol-induced augmentation of Fos in the caudate-putamen and nucleus accumbens of Wistar rats. Haloperidol 68-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 10332800-5 1999 Fos-immunoreactive cells appear in large numbers in all parts of the striatum 3 h after the administration of haloperidol. Haloperidol 110-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 10332800-6 1999 Pretreatment with MK801 attenuates the haloperidol-induced increase in Fos in the caudate-putamen. Dizocilpine Maleate 18-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 10332800-6 1999 Pretreatment with MK801 attenuates the haloperidol-induced increase in Fos in the caudate-putamen. Haloperidol 39-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 10560537-0 1999 Xylene-induced effects on brain neurotransmitters, behavior and fos protein in rats. Xylenes 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 10560537-4 1999 On the contrary, xylene at a dose of 500 mg/kg increased the beta-endorphin level in caudate and c-fos expression in hippocampus. Xylenes 17-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 10560537-5 1999 These data suggest that the xylene-induced behavioral alterations might be associated with the expression of Fos protein in the hippocampus. Xylenes 28-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-112 10435781-6 1999 Nuclear immunolabeling for the transcription factor, Fos, was observed in several preoptic and hypothalamic sites following 2DG administration. Deoxyglucose 124-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 10401552-0 1999 Non-NMDA glutamate receptors modulate capsaicin induced c-fos expression within trigeminal nucleus caudalis. Capsaicin 38-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 10401552-5 1999 The number of positive c-fos cells increased by 17 fold after intracisternal capsaicin (5 nmol) administration. Capsaicin 77-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 10401552-9 1999 Pretreatment with L-AP4 (1, 3 and 10 mg kg-1) decreased the number of Sp5C c-fos LI cells by a maximum of 30%, whereas GAMS (1 and 10 mg kg-1) and NS-102 (1 and 5 mg kg-1) did not show any significant effect. 2-amino-4-phosphono-propinate 18-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 10435781-7 1999 Rats pretreated with the mu antagonist exhibited significantly fewer Fos-positive neurons in the medial preoptic area and dorsomedial hypothalamic nucleus in response to 2DG, compared to vehicle-pretreated controls. Deoxyglucose 170-173 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 10457601-12 1999 The prevention of cystitis by mesna was accompanied only by a reduction in spinal Fos activity, the supraspinal activities remaining high and in strict relationship with the vagal afferent activity. Mesna 30-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 10350256-6 1999 Adenosine triphosphate and hemoglobin, possible spasmogens present in hemolysate, caused much smaller and more rapid increases in c-fos expression than whole hemolysate. triphosphoric acid 10-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 10336728-5 1999 A single injection of citalopram (10 mg/kg, s.c.), increased the plasma levels of ACTH and corticosterone in a dose-dependent manner and increased the number of c-Fos containing cells in the hypothalamic paraventricular nucleus. Citalopram 22-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 10408970-0 1999 Thymulin induces c-fos expression in the spinal cord of rats which is reversed by meloxicam and morphine. Meloxicam 82-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 10336529-6 1999 Both intracellular Ca2+ chelation (using BAPTA/AM) and extracellular Ca2+ depletion (using EGTA) significantly inhibited PE-induced c-fos expression by alpha1A and alpha1B receptors. 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid 41-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 10336529-6 1999 Both intracellular Ca2+ chelation (using BAPTA/AM) and extracellular Ca2+ depletion (using EGTA) significantly inhibited PE-induced c-fos expression by alpha1A and alpha1B receptors. Egtazic Acid 91-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 10336529-8 1999 The calmodulin (CaM) antagonists, R24571, W7, and trifluoperazine, but not the CaM-dependent protein kinases inhibitor KN-62, significantly inhibited c-fos induction by alpha1A and alpha1B receptors. calmidazolium 34-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-155 10336529-8 1999 The calmodulin (CaM) antagonists, R24571, W7, and trifluoperazine, but not the CaM-dependent protein kinases inhibitor KN-62, significantly inhibited c-fos induction by alpha1A and alpha1B receptors. Trifluoperazine 50-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-155 10435392-5 1999 METHODS: We examined the effects of clozapine pretreatment of rats on haloperidol-elicited forebrain Fos expression, using both immunoblot and immunohistochemical methods. Clozapine 36-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-104 10435392-5 1999 METHODS: We examined the effects of clozapine pretreatment of rats on haloperidol-elicited forebrain Fos expression, using both immunoblot and immunohistochemical methods. Haloperidol 70-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-104 10408246-5 1999 administration of morphine (3 mg/kg) decreased the total number per section of Fos-Like-Immunoreactive (Fos-LI) neurons by 51%, observed at 2 h after injection of carrageenin. Morphine 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 10408246-5 1999 administration of morphine (3 mg/kg) decreased the total number per section of Fos-Like-Immunoreactive (Fos-LI) neurons by 51%, observed at 2 h after injection of carrageenin. Morphine 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 10408246-5 1999 administration of morphine (3 mg/kg) decreased the total number per section of Fos-Like-Immunoreactive (Fos-LI) neurons by 51%, observed at 2 h after injection of carrageenin. Carrageenan 163-174 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 10350530-3 1999 Psychostimulants, which increase dopamine levels, induce Fos-Li in the striatum through D1 receptors. Dopamine 33-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 10350549-0 1999 Anteroventral third ventricle (AV3V) lesions alter c-fos expression induced by salt loading. Salts 79-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 10350549-2 1999 Salt loading in AV3VX rats resulted in reduced but not completely abolished c-fos expression in the supraoptic and paraventricular nuclei. Salts 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 10350549-4 1999 These regions showed c-fos expression following salt loading. Salts 48-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 10377372-0 1999 An antisense oligonucleotide reverses the footshock-induced expression of fos in the rat medial prefrontal cortex and the subsequent expression of conditioned fear-induced immobility. Oligonucleotides 13-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 10377372-8 1999 In other animals, an antisense oligonucleotide directed against the c-fos mRNA was injected into the infralimbic/prelimbic cortex 12 or 72 hr before the acquisition session. Oligonucleotides 31-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 10377372-11 1999 The antisense oligonucleotide blockade of Fos production during acquisition was associated with a significantly less fearful response during the extinction session. Oligonucleotides 14-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 10408970-0 1999 Thymulin induces c-fos expression in the spinal cord of rats which is reversed by meloxicam and morphine. Morphine 96-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 10234044-4 1999 GBR-12935 also produced a reduction in noxious stimulus-induced c-fos expression in nociceptive areas of the spinal dorsal horn, suggesting that dopamine in the RAIC acts in part through descending antinociception. Dopamine 145-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 10320705-0 1999 Time dependence and role of N-methyl-D-aspartate glutamate receptors in the priming of D2-mediated rotational behavior and striatal Fos expression in 6-hydroxydopamine lesioned rats. Oxidopamine 150-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-135 10320705-2 1999 Previously, we have shown that 6-OHDA rats primed with three injections of the D1/D2 dopamine agonist apomorphine (0.5 mg/kg) permitted a challenge with an otherwise inactive dose of the D2 agonist quinpirole (0.25 mg/kg) to elicit robust rotational behavior and to induce Fos expression in striatoentopeduncular neurons. Oxidopamine 31-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 273-276 10320705-2 1999 Previously, we have shown that 6-OHDA rats primed with three injections of the D1/D2 dopamine agonist apomorphine (0.5 mg/kg) permitted a challenge with an otherwise inactive dose of the D2 agonist quinpirole (0.25 mg/kg) to elicit robust rotational behavior and to induce Fos expression in striatoentopeduncular neurons. Apomorphine 102-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 273-276 10320705-4 1999 The enhanced rotational behavior and striatal Fos expression observed following challenge with quinpirole (0.25 mg/kg) peaked 1 day following the third apomorphine priming injection and persisted, in reduced form, for at least 4 months. Quinpirole 95-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 10320705-5 1999 Pretreatment with the NMDA antagonists MK-801 or 3-[(+)-2-carboxypiperazin-4-yl]-propyl-1-phosphonate (CPP) dose-dependently attenuated apomorphine-priming of quinpirole-mediated rotational behavior and striatal Fos induction compared to 6-OHDA rats primed with apomorphine alone. N-Methylaspartate 22-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 212-215 10320705-5 1999 Pretreatment with the NMDA antagonists MK-801 or 3-[(+)-2-carboxypiperazin-4-yl]-propyl-1-phosphonate (CPP) dose-dependently attenuated apomorphine-priming of quinpirole-mediated rotational behavior and striatal Fos induction compared to 6-OHDA rats primed with apomorphine alone. Dizocilpine Maleate 39-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 212-215 10320705-5 1999 Pretreatment with the NMDA antagonists MK-801 or 3-[(+)-2-carboxypiperazin-4-yl]-propyl-1-phosphonate (CPP) dose-dependently attenuated apomorphine-priming of quinpirole-mediated rotational behavior and striatal Fos induction compared to 6-OHDA rats primed with apomorphine alone. 3-(2-carboxypiperazin-4-yl)propyl-1-phosphonic acid 49-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 212-215 10320705-5 1999 Pretreatment with the NMDA antagonists MK-801 or 3-[(+)-2-carboxypiperazin-4-yl]-propyl-1-phosphonate (CPP) dose-dependently attenuated apomorphine-priming of quinpirole-mediated rotational behavior and striatal Fos induction compared to 6-OHDA rats primed with apomorphine alone. 3-(2-carboxypiperazin-4-yl)propyl-1-phosphonic acid 103-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 212-215 10399540-2 1999 We investigated the c-Fos-like immunoreactivity of the brainstem and upper cervical spinal cord (C1 region) following an injection of mustard oil (15 microliters of 20%) into the nasal mucosa of pentobarbital anesthetized rats after exposure to hyperbaric (2-atmospheres, 1 h) and normobaric pressures. mustard oil 134-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 10399540-2 1999 We investigated the c-Fos-like immunoreactivity of the brainstem and upper cervical spinal cord (C1 region) following an injection of mustard oil (15 microliters of 20%) into the nasal mucosa of pentobarbital anesthetized rats after exposure to hyperbaric (2-atmospheres, 1 h) and normobaric pressures. Pentobarbital 195-208 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 10494488-0 1999 Variations of progesterone receptor and c-fos gene expression in the rat uterus after treatment with norethisterone and its A-ring reduced metabolites. Norethindrone 101-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 10494488-2 1999 The aim of this work was to investigate the estrogenic properties of norethisterone (NET) and its A-ring-reduced derivatives by determining progesterone receptor (PR) and c-fos mRNA content of two estrogen-regulated genes in the uterus of ovariectomized rats. Norethindrone 69-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-176 10494488-4 1999 The highest PR and c-fos mRNA content was observed 3 h and 2 h after 17 beta-estradiol administration, respectively. Estradiol 72-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 10494488-7 1999 The increase in c-fos mRNA content induced by these reduced compounds was lower than that found with estradiol treatment. Estradiol 101-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 10386521-7 1999 c-fos activation after kainic acid injection was not observed in the region of the dysplasia. Kainic Acid 23-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 10319324-4 1999 Moreover, cAMP-induced c-fos promoter activity is decreased in the somatolactotroph cell line GH3 when Pit-1 expression is reduced by hybrid arrest with an antisense sequence complementary to Pit-1 cDNA. Cyclic AMP 10-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 10353503-7 1999 The number of Fos stained cells in the dorsal horn of the spinal cord, induced by the formalin treatment, was decreased significantly by the 3.0 mg dose of morphine at all three ages. Formaldehyde 86-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 10320729-2 1999 In this study, we have examined the effect of drugs which spontaneously release NO (sodium nitroprusside, SNP) or inhibit the NO synthase (NOS) enzyme (Nomega-nitro-L-arginine, L-NA) on the activity of some hypothalamic and functionally associated nuclei using Fos expression as an index of neuronal activation. Nitroarginine 152-175 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 261-264 10320729-5 1999 Urethane-anesthetized animals, compared with their conscious counterparts, showed increased Fos expression in the PVN and SON. Urethane 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 10320734-6 1999 Expression of immediate early genes (IEGs; c-fos and NGFI-A) was observed in the cortex, thalamic nuclei, subthalamic nucleus, pars reticulata of the substantia nigra, lateral and medial geniculate bodies on the ipsilateral side from 3 to 15 h after FeCl2 injection. ferrous chloride 250-255 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 10216194-5 1999 Neural activity was characterized using immunohistochemistry to detect the immediate early gene product Fos in serotonin-immunoreactive cells in the DRN. Serotonin 111-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 10363926-3 1999 Fos expression was increased in the ARC after capsaicin injection compared with vehicle-treated rats. Capsaicin 46-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 10363926-4 1999 Pretreatment with the NO synthase (NOS) inhibitor, N(omega)-nitro-L-arginine methyl ester (L-NAME) attenuated the effect of capsaicin on Fos expression and NADPH-d reactivity in the ARC-ME in comparison with rats injected with D-NAME, the inactive stereoisomer of L-NAME. NG-Nitroarginine Methyl Ester 51-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-140 10363926-4 1999 Pretreatment with the NO synthase (NOS) inhibitor, N(omega)-nitro-L-arginine methyl ester (L-NAME) attenuated the effect of capsaicin on Fos expression and NADPH-d reactivity in the ARC-ME in comparison with rats injected with D-NAME, the inactive stereoisomer of L-NAME. NG-Nitroarginine Methyl Ester 91-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-140 10363926-4 1999 Pretreatment with the NO synthase (NOS) inhibitor, N(omega)-nitro-L-arginine methyl ester (L-NAME) attenuated the effect of capsaicin on Fos expression and NADPH-d reactivity in the ARC-ME in comparison with rats injected with D-NAME, the inactive stereoisomer of L-NAME. Capsaicin 124-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-140 10216178-0 1999 Compartmental changes in expression of c-Fos and FosB proteins in intact and dopamine-depleted striatum after chronic apomorphine treatment. Dopamine 77-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 10216178-0 1999 Compartmental changes in expression of c-Fos and FosB proteins in intact and dopamine-depleted striatum after chronic apomorphine treatment. Apomorphine 118-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 10395074-3 1999 Dexamethasone-dependent stimulation of c-fos and c-jun was modulated predominantly at the level of transcription. Dexamethasone 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 10395074-4 1999 Sustained levels of induced c-fos and c-jun proteins were observed after dexamethasone treatment. Dexamethasone 73-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 10334505-2 1999 The intraplantar injection of carrageenin-induced the development of a peripheral oedema, associated with an increase in Fos-like immunoreactivity at the level of the dorsal horn of the spinal cord. Carrageenan 30-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-124 10334505-8 1999 Pre-treatment with U-69,593 strongly decreased the number of Fos-like Immunoreactive neurones of the spinal cord in a dose-dependent, antagonist reversible manner; maximal effect was 65%. u-69 19-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 10187833-5 1999 In these studies we confirmed that H7, but not HA1004, potently blocks the induction of zif268 and c-fos mRNA by nerve growth factor, carbachol, phorbol ester, Ca2+ ionophore, or forskolin. Carbachol 134-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 10187833-5 1999 In these studies we confirmed that H7, but not HA1004, potently blocks the induction of zif268 and c-fos mRNA by nerve growth factor, carbachol, phorbol ester, Ca2+ ionophore, or forskolin. Phorbol Esters 145-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 10095048-1 1999 morphine on carrageenin evoked c-Fos expression in the superficial dorsal horn of the rat spinal cord. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 10095048-1 1999 morphine on carrageenin evoked c-Fos expression in the superficial dorsal horn of the rat spinal cord. Carrageenan 12-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 10353503-7 1999 The number of Fos stained cells in the dorsal horn of the spinal cord, induced by the formalin treatment, was decreased significantly by the 3.0 mg dose of morphine at all three ages. Morphine 156-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 10321456-0 1999 The effect of lithium on methamphetamine-induced regional Fos protein expression in the rat brain. Lithium 14-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 10321456-0 1999 The effect of lithium on methamphetamine-induced regional Fos protein expression in the rat brain. Methamphetamine 25-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 10321456-3 1999 To investigate the neurobiologic substrates of the antimanic effects of chronic lithium administration, we investigated its effects on methamphetamine-induced regional Fos protein expression in the rat brain. Lithium 80-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-171 10321456-3 1999 To investigate the neurobiologic substrates of the antimanic effects of chronic lithium administration, we investigated its effects on methamphetamine-induced regional Fos protein expression in the rat brain. Methamphetamine 135-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-171 10092632-7 1999 In CS-54 cells, we found that NO increased fos promoter activity and that the increase was prevented by a guanylate cyclase inhibitor. Cesium 3-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 10320002-4 1999 In the rats which were subcutaneously injected with formalin into the upper and lower lips, c-fos protein (Fos) was found in PPD-LI neurons which were labeled with a retrograde tracer injected into the thalamic regions. Formaldehyde 52-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-105 10320002-4 1999 In the rats which were subcutaneously injected with formalin into the upper and lower lips, c-fos protein (Fos) was found in PPD-LI neurons which were labeled with a retrograde tracer injected into the thalamic regions. Formaldehyde 52-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 10198401-0 1999 Distribution of Fos immunoreactivity in rat brain after sodium consumption induced by peritoneal dialysis. Sodium 56-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 10198401-1 1999 Fos immunoreactivity was used to map the neuronal population groups activated after sodium ingestion induced by peritoneal dialysis (PD) in rats. Sodium 84-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 10198401-3 1999 Sodium ingestion stimulated by PD produced Fos immunoreactivity within defined cells groups of the lamina terminalis and hindbrain areas such us the nucleus of the solitary tract, area postrema, and lateral parabrachial nucleus. Sodium 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 10451223-10 1999 The numbers of both Fos/FRA and (Fos/FRA)/TH dual-staining neurons increased in the arcuate nucleus following NAL treatment (p < 0.05 for both comparisons). Naloxone 110-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 10095048-2 1999 This study performed in freely moving rats evaluated the ability of specific opioid receptor antagonists to reverse the inhibitory effects of morphine on carrageenin-induced c-Fos expression in the spinal cord. Morphine 142-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 174-179 10095048-2 1999 This study performed in freely moving rats evaluated the ability of specific opioid receptor antagonists to reverse the inhibitory effects of morphine on carrageenin-induced c-Fos expression in the spinal cord. Carrageenan 154-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 174-179 10095048-6 1999 morphine (3 mg/kg) produced a marked decrease (58+/-5%) in the number of Fos-LI neurones measured at 2 h after intraplantar (i.pl.) Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-76 10095048-8 1999 This decrease in c-Fos expression was completely blocked by combined administration of morphine with the mu-opioid receptor antagonist, [D-Phe-Cys-Tyr-D-Orn-Thr-Pen-Thr-NH2] (CTOP-1+1 mg/kg). Morphine 87-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 10095048-8 1999 This decrease in c-Fos expression was completely blocked by combined administration of morphine with the mu-opioid receptor antagonist, [D-Phe-Cys-Tyr-D-Orn-Thr-Pen-Thr-NH2] (CTOP-1+1 mg/kg). [d-phe-cys-tyr-d-orn-thr-pen-thr-nh2 136-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 10451223-10 1999 The numbers of both Fos/FRA and (Fos/FRA)/TH dual-staining neurons increased in the arcuate nucleus following NAL treatment (p < 0.05 for both comparisons). Naloxone 110-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 10460009-7 1999 Furthermore, this c-fos expression is not inhibited by cycloheximide, but is completely abolished by pretreatment with TPA, so that the c-fos gene is a direct target of TGF-beta1 signalling and PKC is involved in this c-fos induction. Cycloheximide 55-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 10460009-7 1999 Furthermore, this c-fos expression is not inhibited by cycloheximide, but is completely abolished by pretreatment with TPA, so that the c-fos gene is a direct target of TGF-beta1 signalling and PKC is involved in this c-fos induction. Tetradecanoylphorbol Acetate 119-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 10460009-7 1999 Furthermore, this c-fos expression is not inhibited by cycloheximide, but is completely abolished by pretreatment with TPA, so that the c-fos gene is a direct target of TGF-beta1 signalling and PKC is involved in this c-fos induction. Tetradecanoylphorbol Acetate 119-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 10216178-2 1999 In order to understand striatal mechanisms behind this long-term behavioral change we examined striatal c-Fos and FosB immunoreactivity induced by apomorphine challenge (5 mg/kg, s.c.) after 3 days of withdrawal following a 2-week administration (5 mg/kg, b.i.d., s.c.) both in intact and 6-OHDA-lesioned animals. Apomorphine 147-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 10460009-7 1999 Furthermore, this c-fos expression is not inhibited by cycloheximide, but is completely abolished by pretreatment with TPA, so that the c-fos gene is a direct target of TGF-beta1 signalling and PKC is involved in this c-fos induction. Tetradecanoylphorbol Acetate 119-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 10216178-3 1999 In intact rats, c-Fos induction by acute apomorphine exposure showed a striosomal pattern, whereas FosB immunopositivity was diffusely distributed. Apomorphine 41-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 10194650-0 1999 Serotonin agonist quipazine induces photic-like phase shifts of the circadian activity rhythm and c-Fos expression in the rat suprachiasmatic nucleus. Serotonin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 10194650-0 1999 Serotonin agonist quipazine induces photic-like phase shifts of the circadian activity rhythm and c-Fos expression in the rat suprachiasmatic nucleus. Quipazine 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 10194650-5 1999 More important, quipazine induced significant phase advances of the activity rhythm and c-Fos production in the SCN at the end of the subjective night (Circadian Time [CT] 22), whereas neither phase shifts nor c-Fos induction were observed during the subjective day. Quipazine 16-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 11939005-0 1999 [Effect of dimethoate on the expression of c-fos gene in skeletal muscle]. Dimethoate 11-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 10098858-6 1999 In drug-naive animals, acute amphetamine induced the expression of RGS2, 3, and 5 and the immediate early genes c-fos and zif/268. Amphetamine 29-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 11499015-0 1999 [Expression of c-fos in the medulla oblongata after carotid baroreceptor activation by elevated intrasinus pressure and adenosine]. Adenosine 120-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 11499015-1 1999 Expression of c-fos protein in the medulla oblongata after baroreceptor activation by elevated intrasinus pressure (ISP) and perfusion of adenosine (Ado) was examined in 14 vascularly isolated carotid sinus perfusion rats. Adenosine 138-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 11499015-1 1999 Expression of c-fos protein in the medulla oblongata after baroreceptor activation by elevated intrasinus pressure (ISP) and perfusion of adenosine (Ado) was examined in 14 vascularly isolated carotid sinus perfusion rats. Adenosine 149-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 11939005-2 1999 The result showed that c-fos mRNA and protein were significantly increased in skeletal muscle of rat after dosing with dimethoate. Dimethoate 119-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 11939005-3 1999 These results indicated that c-fos might act as transcription factor and regulate other gene expression during the early period of organophosphate intoxication. Organophosphates 131-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 10225363-3 1999 To establish the relative importance of these subtypes, we compared the effects of sumatriptan with those of a selective 5-HT1F receptor agonist (LY 344864) on c-fos protein expression in the trigeminal nucleus caudalis. LY 344864 146-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 10225363-4 1999 c-fos expression was induced in urethane-anaesthetized rats by intracisternal capsaicin administration. Urethane 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 10225363-4 1999 c-fos expression was induced in urethane-anaesthetized rats by intracisternal capsaicin administration. Capsaicin 78-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 10225363-5 1999 Sumatriptan and LY 344864 decreased the number of capsaicin-induced c-fos-like immunoreactive cells within trigeminal nucleus caudalis (ID50 = 0.04 and 0.6 mg kg(-1)). Sumatriptan 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 10225363-5 1999 Sumatriptan and LY 344864 decreased the number of capsaicin-induced c-fos-like immunoreactive cells within trigeminal nucleus caudalis (ID50 = 0.04 and 0.6 mg kg(-1)). LY 344864 16-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 10225363-5 1999 Sumatriptan and LY 344864 decreased the number of capsaicin-induced c-fos-like immunoreactive cells within trigeminal nucleus caudalis (ID50 = 0.04 and 0.6 mg kg(-1)). Capsaicin 50-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 10082881-1 1999 It was previously reported that systemic administration of the nonselective opioid antagonist, naltrexone, induces Fos-like immunoreactivity (FLI) within the central nucleus of the amygdala (CeA), bed nucleus of the stria terminalis (lateral-dorsal division; BSTLD), nucleus accumbens shell (NACshell) and ventral tegmental area (VTA) of free-feeding rats. Naltrexone 95-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-118 10225363-7 1999 LY 344864 appears to attenuate c-fos-like immunoreactivity via 5-HT1F receptors, while sumatriptan acts via 5-HT1B receptors. LY 344864 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 10082881-3 1999 Considering the involvement of mesoaccumbens dopamine neurons and components of the "extended amygdala" in motivated behavior and reward, it was hypothesized that the induction of c-Fos by naltrexone accounts for the motivational-affective consequences of opioid antagonism. Dopamine 45-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 180-185 10218911-2 1999 Intrathecal administration of c-fos antisense (30 nmol/20 microl) into the lumbar region (L1-L5) 18 h prior to nerve ligation attenuated 80% of Fos-immunoreactivity 90 min after ligation compared to rats infused with c-fos sense or saline. Sodium Chloride 232-238 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 10082881-3 1999 Considering the involvement of mesoaccumbens dopamine neurons and components of the "extended amygdala" in motivated behavior and reward, it was hypothesized that the induction of c-Fos by naltrexone accounts for the motivational-affective consequences of opioid antagonism. Naltrexone 189-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 180-185 10082881-8 1999 reproduced the effect of naltrexone in BSTLD and CeA, suggesting that the induction of c-Fos in these two structures is a consequence of kappa receptor blockade. Naltrexone 25-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 10082881-10 1999 ), reproduced the effect of naltrexone in NACshell, suggesting that the induction of c-Fos in this structure is a consequence of mu receptor blockade. Naltrexone 28-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 10095079-1 1999 Our previous studies demonstrated that nicotine induces c-fos expression in the suprachiasmatic nucleus (SCN) of the rat during a narrow developmental window occurring in the perinatal period. Nicotine 39-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 10372514-0 1999 Nicotine phase shifts the 6-sulphatoxymelatonin rhythm and induces c-Fos in the SCN of rats. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 10372514-2 1999 This study examined the effect of the cholinergic agonists nicotine and oxotremorine on (1) the rhythmic production of melatonin using the metabolite, 6-sulphatoxymelatonin as a marker, and (2) the expression of c-Fos protein in the suprachiasmatic nuclei (SCN) of the rat. Oxotremorine 72-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 212-217 10372514-4 1999 Nicotine administration also caused the induction of c-Fos-like immunoreactivity in the SCN in a dose- and time-dependent manner. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 10372514-5 1999 Further, pre-treatment with the nicotinic antagonist mecamylamine reduced the number of nicotine-induced c-Fos-positive cells in the SCN by 65%. Mecamylamine 53-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 10372514-5 1999 Further, pre-treatment with the nicotinic antagonist mecamylamine reduced the number of nicotine-induced c-Fos-positive cells in the SCN by 65%. Nicotine 88-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 10780834-0 1999 Effect of nitric oxide synthesis inhibition on c-Fos expression in hippocampus and cerebral cortex following two forms of learning in rats: an immunohistochemistry study. Nitric Oxide 10-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 10064802-4 1999 The mechanisms mediating this neuronal death are unclear though manipulations of dopamine transmission can induce striatal c-fos expression and continuous c-fos expression has been implicated in the molecular cascades controlling apoptosis. Dopamine 81-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 10203239-0 1999 Hypoxia induces c-Fos protein expression in NMDA but not AMPA glutamate receptor labeled neurons within the nucleus tractus solitarii of the conscious rat. N-Methylaspartate 44-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 10203239-3 1999 Hypoxia markedly increased c-Fos immunoreactivity within nTS neurons which in the vast majority co-labeled for NMDA. N-Methylaspartate 111-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 10435037-0 1999 Reactive oxygen species modulate endothelin-I-induced c-fos gene expression in cardiomyocytes. Reactive Oxygen Species 0-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 10190566-4 1999 H2O2 preferentially induced the expression of c-fos, c-jun, c-myc and egr-1, while JunB and JunD levels remained almost unchanged. Hydrogen Peroxide 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 10103112-7 1999 (ii) After challenges that increase DA by depolarizing DAergic neurons (injection stress or 2 mg/kg morphine), the shell presented the largest increase in DA levels and Fos-LI. Morphine 100-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-172 10103112-10 1999 (iv) After a full D1 agonist (SKF82958), Fos-LI was highest in the shell and lowest in the dorsolateral striatum. SK and F 82958 30-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 10103090-1 1999 A remarkable feature of dopamine functioning is that the concomitant activation of D1-like and D2-like receptors acts to intensify the expression of various dopamine-dependent effects, in particular the expression of the immediate-early genes, c-fos and zif268. Dopamine 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 244-249 10103090-1 1999 A remarkable feature of dopamine functioning is that the concomitant activation of D1-like and D2-like receptors acts to intensify the expression of various dopamine-dependent effects, in particular the expression of the immediate-early genes, c-fos and zif268. Dopamine 157-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 244-249 10435037-4 1999 The ROS-inducible c-fos expression was analyzed by Northern blotting and promoter activity. Reactive Oxygen Species 4-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 10435037-8 1999 Et-1 or H2O2 treatment of cardiomyocytes rapidly induced the expression of an immediate early gene c-fos. Hydrogen Peroxide 8-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 10435037-9 1999 Et-1-treated cardiomyocytes enhanced the c-fos gene expression as revealed by functional analysis using a reporter gene construct containing c-fos promoter region (-2.25 kb) and reporter gene chloramphenicol acetyltransferase. Endothelin-1 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 10435037-9 1999 Et-1-treated cardiomyocytes enhanced the c-fos gene expression as revealed by functional analysis using a reporter gene construct containing c-fos promoter region (-2.25 kb) and reporter gene chloramphenicol acetyltransferase. Endothelin-1 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 10435037-10 1999 The induction of mRNA levels and the promoter activities of c-fos gene by Et-1 or H2O2 were abolished by pretreating cardiomyocytes with catalase or NAC. Hydrogen Peroxide 82-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 10435037-10 1999 The induction of mRNA levels and the promoter activities of c-fos gene by Et-1 or H2O2 were abolished by pretreating cardiomyocytes with catalase or NAC. Acetylcysteine 149-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 10435037-15 1999 CONCLUSIONS: These findings clearly indicate that Et-1 treatment to cardiomyocytes can induce ROS via Ras pathway and the increased ROS are involved in the increase of c-fos expression. Reactive Oxygen Species 132-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-173 10094419-8 1999 These data reveal that only discrete populations of NADPH-d-containing neurons in the caudal brainstem are transcriptionally activated via the Fos stimulus-transcription cascade in response to glucose substrate imbalance, and suggest that NO and/or other neurotransmitters released by these neurons may function as neurochemical mediators of glucoprivic regulatory effects within this part of the brain. Glucose 193-200 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-146 10051214-4 1999 However, in the deeper layers of the spinal cord (V-X), there was no such correlation, and stimulation of joint nociceptors and formalin-induced inflammation produced the greatest proportion of Fos/neurokinin-1 co-localization, suggesting a particular role for substance P in the mediation of joint pain and inflammatory hyperalgesia. Formaldehyde 128-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 194-197 10077330-4 1999 Striatal infusion of 6-cyano-7-nitroquinoxaline-2,3-dione (1 mM) completely blocked (+/-)-alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate-induced c-fos and zif268 expression. 6-Cyano-7-nitroquinoxaline-2,3-dione 21-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-156 10077330-4 1999 Striatal infusion of 6-cyano-7-nitroquinoxaline-2,3-dione (1 mM) completely blocked (+/-)-alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate-induced c-fos and zif268 expression. 3-hydroxy-5-methylisoxazole-4-propionate 102-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-156 10077330-9 1999 In contrast, tetrodotoxin abolished induction of c-fos and zif268 messenger RNAs by the D2 antagonist eticlopride (0.5 mg/kg, i.p.) Tetrodotoxin 13-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 10077330-9 1999 In contrast, tetrodotoxin abolished induction of c-fos and zif268 messenger RNAs by the D2 antagonist eticlopride (0.5 mg/kg, i.p.) eticlopride 102-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 10087450-0 1999 Site-specific induction of Fos immunoreactivity in preoptic and hypothalamic NADPH-positive neurons during glucoprivation. NADP 77-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 10087450-2 1999 The following studies examined whether populations of NADPH-d-positive neurons in the hypothalamus and nearby preoptic area express immunoreactivity for the nuclear transcription factor, Fos, in response to glucose substrate imbalance. Glucose 207-214 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 187-190 10087450-7 1999 The glucose antimetabolite elicited expression of nuclear Fos-ir by NADPH-d-positive neurons in several neural structures, including the medial preoptic area, median preoptic nucleus, anterior commissural, periventricular magnocellular supraoptic nucleus, paraventricular nucleus, and medial part of the bed nucleus of the stria terminalis. Glucose 4-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 10199625-3 1999 An increase in cAMP levels generated by these unopposed receptors would then lead to the well-described behavioral and molecular effects of antipsychotic administration such as catalepsy and striatal c-fos and neurotensin gene transcription. Cyclic AMP 15-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 200-205 10103090-3 1999 Pretreatment with SR48692 (3 and 10 mg/kg) reduced the number of Fos-like immunoreactive cells produced by the combined administration of SKF38393 (20 mg/kg) and quinpirole (1 mg/kg) in the caudate-putamen, nucleus accumbens, globus pallidus and ventral pallidum. SR 48692 18-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 10103090-3 1999 Pretreatment with SR48692 (3 and 10 mg/kg) reduced the number of Fos-like immunoreactive cells produced by the combined administration of SKF38393 (20 mg/kg) and quinpirole (1 mg/kg) in the caudate-putamen, nucleus accumbens, globus pallidus and ventral pallidum. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 138-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 10103090-3 1999 Pretreatment with SR48692 (3 and 10 mg/kg) reduced the number of Fos-like immunoreactive cells produced by the combined administration of SKF38393 (20 mg/kg) and quinpirole (1 mg/kg) in the caudate-putamen, nucleus accumbens, globus pallidus and ventral pallidum. Quinpirole 162-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 10103090-5 1999 Pretreatment with SR48692 (10 mg/kg) also diminished Fos induction by the indirect dopamine agonist, cocaine (25 mg/kg), particularly at the rostral level of the caudate-putamen. SR 48692 18-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 10199625-6 1999 Both the specific adenosine A2a antagonist 8-(3-chlorostyryl)-caffeine and the general adenosine antagonist theophylline reduced haloperidol-dependent induction of striatal neurotensin and c-fos messenger RNA. Haloperidol 129-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 189-194 10023034-6 1999 Finally, the induction of Fos-like immunoreactivity (Fos-ir) following either DFEN or amylin was examined in both APX and sham operated groups. Dexfenfluramine 78-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 10023034-6 1999 Finally, the induction of Fos-like immunoreactivity (Fos-ir) following either DFEN or amylin was examined in both APX and sham operated groups. Dexfenfluramine 78-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 10103090-5 1999 Pretreatment with SR48692 (10 mg/kg) also diminished Fos induction by the indirect dopamine agonist, cocaine (25 mg/kg), particularly at the rostral level of the caudate-putamen. Dopamine 83-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 10103090-5 1999 Pretreatment with SR48692 (10 mg/kg) also diminished Fos induction by the indirect dopamine agonist, cocaine (25 mg/kg), particularly at the rostral level of the caudate-putamen. Cocaine 101-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 10194112-0 1999 Acoustic brainstem nuclei express Fos after flurothyl-induced generalized seizures in rats. Flurothyl 44-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 10218785-0 1999 D-Fenfluramine induces serotonin-mediated Fos expression in corticotropin-releasing factor and oxytocin neurons of the hypothalamus, and serotonin-independent Fos expression in enkephalin and neurotensin neurons of the amygdala. Dexfenfluramine 0-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 10218785-0 1999 D-Fenfluramine induces serotonin-mediated Fos expression in corticotropin-releasing factor and oxytocin neurons of the hypothalamus, and serotonin-independent Fos expression in enkephalin and neurotensin neurons of the amygdala. Serotonin 23-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 10218785-0 1999 D-Fenfluramine induces serotonin-mediated Fos expression in corticotropin-releasing factor and oxytocin neurons of the hypothalamus, and serotonin-independent Fos expression in enkephalin and neurotensin neurons of the amygdala. Serotonin 137-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-162 10218785-3 1999 Induction of Fos immunoreactivity following D-fenfluramine injection was used as an index of neuronal activation. Dexfenfluramine 44-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 10218785-7 1999 Fluoxetine pretreatment blocked Fos expression in corticotropin-releasing factor- and oxytocin-expressing cells in the hypothalamus, but not in enkephalin-and neurotensin-expressing cells located in the bed nucleus of the stria terminalis and central amygdaloid nucleus. Fluoxetine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 10218785-11 1999 Induction of Fos expression by D-fenfluramine in restricted populations of cells suggests a selective activation of neuronal circuitry that is likely to be involved in the appetite suppressant effects of D-fenfluramine. Dexfenfluramine 31-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 10218785-11 1999 Induction of Fos expression by D-fenfluramine in restricted populations of cells suggests a selective activation of neuronal circuitry that is likely to be involved in the appetite suppressant effects of D-fenfluramine. Dexfenfluramine 204-218 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 10204725-4 1999 Compared to vehicle-injected animals with injection of vehicle alone, microinjection of a serotoninergic neurotoxin 5,7-dihydroxytryptamine into the NRM significantly increased thermal hyperalgesia and Fos protein expression in lumbar spinal cord after hindpaw inflammation. 5,7-Dihydroxytryptamine 116-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 202-205 10204725-5 1999 In contrast, the selective bilateral destruction of the NGC with a soma-selective excitotoxic neurotoxin, ibotenic acid, led to an attenuation of hyperalgesia and a reduction of inflammation-induced spinal Fos expression. Ibotenic Acid 106-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 206-209 10199625-6 1999 Both the specific adenosine A2a antagonist 8-(3-chlorostyryl)-caffeine and the general adenosine antagonist theophylline reduced haloperidol-dependent induction of striatal neurotensin and c-fos messenger RNA. 8-(3-chlorostyryl)caffeine 43-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 189-194 10199625-6 1999 Both the specific adenosine A2a antagonist 8-(3-chlorostyryl)-caffeine and the general adenosine antagonist theophylline reduced haloperidol-dependent induction of striatal neurotensin and c-fos messenger RNA. Adenosine 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 189-194 10199625-6 1999 Both the specific adenosine A2a antagonist 8-(3-chlorostyryl)-caffeine and the general adenosine antagonist theophylline reduced haloperidol-dependent induction of striatal neurotensin and c-fos messenger RNA. Theophylline 108-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 189-194 9952411-0 1999 Suppression of postischemic hippocampal nerve growth factor expression by a c-fos antisense oligodeoxynucleotide. Oligodeoxyribonucleotides 92-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 10452091-0 1999 Effect of tetrahydropalmatine analogs on Fos expression induced by formalin-pain. tetrahydropalmatine 10-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 10452091-0 1999 Effect of tetrahydropalmatine analogs on Fos expression induced by formalin-pain. Formaldehyde 67-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 10452091-1 1999 AIM: To study the effect of tetrahydropalmatine (THP) analogs on Fos protein expression induced by formalin-pain and elucidate analgesic mechanism of THP analogs. tetrahydropalmatine 28-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 10452091-1 1999 AIM: To study the effect of tetrahydropalmatine (THP) analogs on Fos protein expression induced by formalin-pain and elucidate analgesic mechanism of THP analogs. tetrahydropalmatine 49-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 10452091-1 1999 AIM: To study the effect of tetrahydropalmatine (THP) analogs on Fos protein expression induced by formalin-pain and elucidate analgesic mechanism of THP analogs. Formaldehyde 99-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 10452091-8 1999 Moreover, the Fos protein expression induced by l-THP and spiperone could be prevented by the pre-treatment of the D2 agonist quinpirole but not D1 agonist SKF38393. tetrahydropalmatine 48-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 10452091-8 1999 Moreover, the Fos protein expression induced by l-THP and spiperone could be prevented by the pre-treatment of the D2 agonist quinpirole but not D1 agonist SKF38393. Spiperone 58-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 10452091-8 1999 Moreover, the Fos protein expression induced by l-THP and spiperone could be prevented by the pre-treatment of the D2 agonist quinpirole but not D1 agonist SKF38393. Quinpirole 126-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 10082860-0 1999 Effects of morphine on the distribution of Fos protein in the trigeminal subnucleus caudalis neurons during experimental tooth movement of the rat molar. Morphine 11-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 10082860-2 1999 Furthermore, the effect of morphine and naloxone on the levels of Fos-IR neurons in the SpVc was examined. Naloxone 40-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 10082860-10 1999 significantly reduced the induction of Fos-IR neurons at the superficial layers of the ipsilateral SpVc in a dose-dependent manner, and its effect was antagonized by the subsequent treatment of naloxone (2 mg/kg, i.p.). Naloxone 194-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 10082860-11 1999 Naloxone pretreatment enhanced the expression of Fos-IR neurons on the ipsilateral SpVc. Naloxone 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 10082860-12 1999 The present results of a reduction of Fos-IR neurons by morphine pretreatment suggest that the induction of Fos-IR neurons may be due to the noxious stimulation caused by induction of experimental tooth movement. Morphine 56-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 10082860-12 1999 The present results of a reduction of Fos-IR neurons by morphine pretreatment suggest that the induction of Fos-IR neurons may be due to the noxious stimulation caused by induction of experimental tooth movement. Morphine 56-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 9952411-1 1999 We examined the uptake and distribution of an antisense phosphorothioated oligodeoxynucleotide (s-ODN) to c-fos, rncfosr115, infused into the left cerebral ventricle of male Long-Evans rats and the effect of this s-ODN on subsequent Fos, NGF, neurotrophin-3 (NT-3), and actin expression. phosphorothioated 56-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 9920930-7 1999 Thus, in a physiological context the Ras-Raf-MEK-ERK pathway, but not RhoA, is required for LPA-stimulated c-Fos expression in Rat-1 cells. lysophosphatidic acid 92-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 10082844-0 1999 Role of NMDA glutamate receptors in regulating D2 dopamine-dependent Fos induction in the rat striatopallidal pathway. Dopamine 50-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 10082844-1 1999 Acute administration of reserpine induces Fos expression in striatopallidal neurons, an effect blocked by pretreatment with the D2 dopamine agonist quinpirole. Reserpine 24-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 10082844-1 1999 Acute administration of reserpine induces Fos expression in striatopallidal neurons, an effect blocked by pretreatment with the D2 dopamine agonist quinpirole. Dopamine 131-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 10082844-1 1999 Acute administration of reserpine induces Fos expression in striatopallidal neurons, an effect blocked by pretreatment with the D2 dopamine agonist quinpirole. Quinpirole 148-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 10082844-2 1999 Pretreatment with the NMDA antagonists (+)MK-801 or CPP attenuated reserpine-mediated striatal Fos induction whereas pretreatment with ketamine or the inactive isomer (-)MK-801 did not. N-Methylaspartate 22-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 10082844-2 1999 Pretreatment with the NMDA antagonists (+)MK-801 or CPP attenuated reserpine-mediated striatal Fos induction whereas pretreatment with ketamine or the inactive isomer (-)MK-801 did not. Dizocilpine Maleate 42-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 10082844-2 1999 Pretreatment with the NMDA antagonists (+)MK-801 or CPP attenuated reserpine-mediated striatal Fos induction whereas pretreatment with ketamine or the inactive isomer (-)MK-801 did not. Reserpine 67-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 9928825-4 1999 Immunohistochemistry revealed that Te induces an increased nuclear expression of c-Fos in SCs. Tellurium 35-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 9928825-12 1999 These results support the hypothesis that the depletion of cholesterol induced by Te treatment triggers a stress response in myelinating SCs mediated by immediate early genes of the fos family. Cholesterol 59-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 182-185 9950818-9 1999 There were significant increases in JunD mRNA and protein in DFMO-treated cells, although expression of the c-fos, c-jun, and junB genes decreased. Eflornithine 61-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 10023034-8 1999 DFEN-induced Fos-ir was reduced greatly in the PVN of APX rats, but appeared normal in several other regions surveyed, including the central nucleus of the amygdala and the dorsal striatum. Dexfenfluramine 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 10023034-10 1999 These data indicate that neither the anorexia nor the flavor aversion that are produced by DFEN are dependent upon the AP, and in particular that Fos-ir induced by DFEN in the LPBE is not due to afferents from the AP/NTS. Dexfenfluramine 164-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-149 9920881-3 1999 Fos kinase is a growth factor-stimulated serine/threonine protein kinase that phosphorylates c-Fos at serine 362 within the carboxyl-terminal regulatory domain. Serine 41-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9920881-3 1999 Fos kinase is a growth factor-stimulated serine/threonine protein kinase that phosphorylates c-Fos at serine 362 within the carboxyl-terminal regulatory domain. Serine 41-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 9920881-5 1999 We have purified Fos kinase by affinity chromatography using the Sepharose-linked protein kinase inhibitor, bisindolylmaleimide (BIM). bisindolylmaleimide 108-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 9920881-5 1999 We have purified Fos kinase by affinity chromatography using the Sepharose-linked protein kinase inhibitor, bisindolylmaleimide (BIM). bisindolylmaleimide 129-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 9920930-1 1999 Lysophosphatidic acid (LPA) stimulates the c-Fos serum response element (SRE) by activating two distinct signal pathways regulated by the small GTPases, Ras and RhoA. lysophosphatidic acid 0-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 9920930-1 1999 Lysophosphatidic acid (LPA) stimulates the c-Fos serum response element (SRE) by activating two distinct signal pathways regulated by the small GTPases, Ras and RhoA. lysophosphatidic acid 23-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 9920930-5 1999 Pertussis toxin and PD98059 strongly inhibited LPA-stimulated c-Fos expression and activation of a SRE:Luc reporter. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 20-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 9889345-0 1999 Differential decreases in c-fos and aldolase C mRNA expression in the rat cerebellum after repeated administration of methamphetamine. Methamphetamine 118-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 11972170-4 1999 Protein kinase C(PKC) agonist PMA induced c-fos expression.PKC inhibitor staurosporine blocked ET-1 induced c-fos expression. Staurosporine 73-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 11972170-4 1999 Protein kinase C(PKC) agonist PMA induced c-fos expression.PKC inhibitor staurosporine blocked ET-1 induced c-fos expression. Staurosporine 73-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 11972177-2 1999 The results showed that subcutaneous injection of formalin in the right hindpaw increased c-fos-like immunoreactivity (FLI), somatostatin-like immunoreactivity (Som-LI), Som-LI/FLI and perprosomatostatin mRNA (PPS-mRNA) in neurones of right spinal dorsal horn and significantly enhanced pain intensity rating. Formaldehyde 50-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 11972177-4 1999 The decrease of c-fos or Som level due to intrathecal injection of ACTH in rats with chronic pain was prevented by injection of cyproheptadine, but not by bicuculline and naloxone. Cyproheptadine 128-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 10096431-0 1999 c-fos gene expression in rat liver is induced by phenobarbital. Phenobarbital 49-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 10096431-1 1999 In the present study we investigated c-fos expression in rat livers, that was initiated with the three arylamines, 2-acetylaminofluorene, 2-acetylaminophenanthrene and trans-4-acetylaminostilbene. aniline 103-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 10096431-1 1999 In the present study we investigated c-fos expression in rat livers, that was initiated with the three arylamines, 2-acetylaminofluorene, 2-acetylaminophenanthrene and trans-4-acetylaminostilbene. 2-Acetylaminofluorene 115-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 10096431-1 1999 In the present study we investigated c-fos expression in rat livers, that was initiated with the three arylamines, 2-acetylaminofluorene, 2-acetylaminophenanthrene and trans-4-acetylaminostilbene. 2-acetamidophenanthrene 138-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 10096431-1 1999 In the present study we investigated c-fos expression in rat livers, that was initiated with the three arylamines, 2-acetylaminofluorene, 2-acetylaminophenanthrene and trans-4-acetylaminostilbene. 4-acetylaminostilbene 168-195 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 10096431-3 1999 Gene expression, determined by the mRNA level, and FOS protein were increased after 52 weeks of treatment in arylamine initiated as well as in phenobarbital only treated animals. aniline 109-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 10096431-3 1999 Gene expression, determined by the mRNA level, and FOS protein were increased after 52 weeks of treatment in arylamine initiated as well as in phenobarbital only treated animals. Phenobarbital 143-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 10096431-4 1999 Expression of c-fos seems to be a phenobarbital induced effect that is independent of additional initiator treatments. Phenobarbital 34-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 10096431-6 1999 The results indicate that phenobarbital, a widely used tumor promoter, induces c-fos expression. Phenobarbital 26-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 9878854-5 1999 After UL with floccular l-NAME infusions, Fos expression appeared in bilateral medial vestibular (MVe) and prepositus hypoglossal (PrH) nuclei. NG-Nitroarginine Methyl Ester 24-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 9878854-6 1999 After UL with floccular saline infusions, however, Fos expression was observed only in the ipsi-MVe and contra-PrH. Sodium Chloride 24-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 10027541-0 1999 Noxious thermal stimulation of c-fos activity induced in rat lumbar spinal cord is reduced by AP-5 but not by glycine. 2-amino-5-phosphopentanoic acid 94-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 10027541-1 1999 The effects of intrathecal administration of NMDA (N-methyl-D-aspartic acid) receptor antagonist AP-5 (2-Amino-5-phosphonopentanoic acid), a competitive and specific NMDA antagonist, and glycine on the neuronal expression of c-fos protein (Fos) in the dorsal neurons lumbar segments four and five were studied after noxious heat stimulation. 2-Amino-5-phosphonovalerate 103-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 225-238 10027541-7 1999 This study suggests that excitatory amino acids, e.g. glutamate but not the inhibitory aminos acid, glycine, plays a role in thermal nociception which in turn is mediated, in part, by c-fos activity. Excitatory Amino Acids 25-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 184-189 10027541-7 1999 This study suggests that excitatory amino acids, e.g. glutamate but not the inhibitory aminos acid, glycine, plays a role in thermal nociception which in turn is mediated, in part, by c-fos activity. Glutamic Acid 54-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 184-189 9972767-0 1999 Pre- versus postinjury effects of intravenous GABAergic anesthetics on formalin-induced Fos immunoreactivity in the rat spinal cord. Formaldehyde 71-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 9972767-15 1999 We measured spinal Fos protein after rats received anesthetics before versus after a formalin injection. Formaldehyde 85-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 10064100-8 1999 Other immediate early response genes, c-fos and junB, were also induced following glucose stimulation with kinetics similar to egr-1, whereas c-jun and junD expression were not affected. Glucose 82-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 10079703-6 1999 Recent studies have demonstrated that chronic alcohol intake can reduce hepatic retinoic acid concentrations, diminish retinoid signaling, and enhance activator protein-1 (AP-1 (c-Jun and c-Fos)) expression in rat liver. Alcohols 46-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 188-193 9889323-4 1999 Pretreatment of the SFO with either captopril, an ANG converting enzyme inhibitor, or losartan, an AT1 receptor antagonist, abolished both drinking and Fos-ir induced by ANG I. Captopril 36-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-155 9889323-4 1999 Pretreatment of the SFO with either captopril, an ANG converting enzyme inhibitor, or losartan, an AT1 receptor antagonist, abolished both drinking and Fos-ir induced by ANG I. Losartan 86-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-155 9889323-5 1999 Water intake partially decreased ANG I-induced Fos-ir in the SON and PVN, but not in the other areas. Water 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 10437171-10 1999 CONCLUSION: The formaldehyde-evoked c-fos expression, Som, and Som synthesis of spinal cord were suppressed by Cor through the serotonin receptor of raphe nuclei. Formaldehyde 16-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 9878890-3 1999 After 30 days of CIH exposure in rats, increased c-fos labeling was seen in the nucleus of the solitary tract and ventrolateral medulla as well as other brainstem regions involved in regulation of SND. cih 17-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 9878890-4 1999 Increased expression of c-fos after CIH may indicate changes in neuronal genetic transcription which ultimately modulate SND. cih 36-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 9878676-1 1999 Water deprivation induces expression of the immediate early gene c-fos in specific brain regions, most likely as a result of the activation of cells that are responsive to changes in osmolality and/or blood volume. Water 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 9889345-2 1999 A single dose of methamphetamine induced c-fos mRNA expression in granule and Purkinje cells of both anterior and posterior lobes. Methamphetamine 17-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 9889345-4 1999 Repeated methamphetamine injections reduced methamphetamine-induced c-fos mRNA signals in the anterior hemisphere and in part of the posterior vermis (lobule VII) and posterior hemisphere. Methamphetamine 9-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 9889345-4 1999 Repeated methamphetamine injections reduced methamphetamine-induced c-fos mRNA signals in the anterior hemisphere and in part of the posterior vermis (lobule VII) and posterior hemisphere. Methamphetamine 44-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 9889345-5 1999 Aldolase C mRNA signals in Purkinje cells decreased only in lobules where methamphetamine-induced c-fos signals were not reduced (lobules VI and IX). Methamphetamine 74-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 9889345-6 1999 Therefore, differential decreases in c-fos mRNA and aldolase C mRNA expression after repeated methamphetamine administration depend upon the localization of Purkinje cells in the cerebellum. Methamphetamine 94-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 9889345-7 1999 Since c-fos mRNA and aldolase C mRNA expressions are markers of excitability and the metabolic state of Purkinje cells, respectively, hypofunction of inhibitory Purkinje cells could be induced if methamphetamine is repeatedly injected. Methamphetamine 196-211 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 6-11 9888869-9 1999 The Ang II type 1 (AT1)-receptor blocker irbesartan completely blocked DNA synthesis, migration, MAPK activation, and c-fos induction by Ang II in VSMCs. Irbesartan 41-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 10625069-16 1999 Dehydroepiandrosterone implants had no clear-cut effects on any immunostaining following acute stress, though there was a trend towards lessened adaptation of the Fos response in the septum after steroid treatment. Steroids 196-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 10076920-6 1999 Moreover, pretreatment with candesartan diminished an increase in phenylalanine incorporation, MAP kinase activity, and c-fos gene expression induced by the stretching of cardiomyocytes. candesartan 28-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 9887185-3 1999 RVO induced Fos immunoreactivity in neurons in the DRG of spinal segments T8-L2 on the side ipsilateral to that of occlusion. rvo 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-15 9987016-4 1999 The number of superficial (laminae I-II) and deep (laminae III-VI) spinal dorsal horn cells expressing the c-fos proto-oncogene 2 h after subcutaneous injection of formalin was reduced by 34% and 50%, respectively, in animals with an ipsilateral DRt lesion as compared to non-lesioned rats. Formaldehyde 164-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 10838433-9 1999 Of the compounds tested, only PTH, prostaglandin E(2), 8-bromo-cAMP, and forskolin induced c-fos mRNA levels, indicating that this assay was specific for compounds that are known to induce cAMP and stimulate bone growth. Dinoprostone 35-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 10838433-9 1999 Of the compounds tested, only PTH, prostaglandin E(2), 8-bromo-cAMP, and forskolin induced c-fos mRNA levels, indicating that this assay was specific for compounds that are known to induce cAMP and stimulate bone growth. 8-Bromo Cyclic Adenosine Monophosphate 55-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 10838433-9 1999 Of the compounds tested, only PTH, prostaglandin E(2), 8-bromo-cAMP, and forskolin induced c-fos mRNA levels, indicating that this assay was specific for compounds that are known to induce cAMP and stimulate bone growth. Colforsin 73-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 10838433-9 1999 Of the compounds tested, only PTH, prostaglandin E(2), 8-bromo-cAMP, and forskolin induced c-fos mRNA levels, indicating that this assay was specific for compounds that are known to induce cAMP and stimulate bone growth. Cyclic AMP 63-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 10576589-4 1999 The present study examined the effect of polyamine deprivation on pain-related behaviors and spinal c-fos expression evoked in the formalin test presumed to better reflect clinical pain, using morphine as analgesia control. Formaldehyde 131-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 10576589-6 1999 In addition this treatment prevented the antinociceptive effect of morphine both on behavioral responses and on spinal c-fos expression. Morphine 67-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 10576589-7 1999 In polyamine-deprived rats, despite morphine injection, nociceptive scores remained dramatically high during the intermediate and the late phases of the response and the number of Fos immunoreactive neurons remained largely higher in deeper layers than in morphine control rats. Polyamines 3-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 180-183 9858538-0 1999 Glutamate induces phosphorylation of Elk-1 and CREB, along with c-fos activation, via an extracellular signal-regulated kinase-dependent pathway in brain slices. Glutamic Acid 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 9858538-6 1999 To investigate biochemically the signaling pathways targeting Elk-1 and c-fos in mature neurons, we used a semi-in vivo system composed of brain slices stimulated with the excitatory neurotransmitter glutamate. Glutamic Acid 200-209 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 9858538-13 1999 These data indicate that ERK functions as a common component in two signaling pathways (ERK/Elk-1 and ERK/?/CREB) converging on the c-fos promoter in postmitotic neuronal cells and that CaM-Ks act as positive regulators of these pathways. cam-ks 186-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 9858557-4 1999 c-Fos, c-Jun, and JunB are induced rapidly in response to LPA stimulation, whereas Fra-1 and Fra-2 are induced after a significant lag. lysophosphatidic acid 58-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9858557-6 1999 The early expression of c-Fos can be reconstituted with nonmitogenic doses of LPA, but the response is transient compared to that observed with mitogenic doses. lysophosphatidic acid 78-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 9858557-8 1999 LPA-stimulated expression of c-Fos, Fra-1, Fra-2, c-Jun, and JunB is inhibited by the MEK1 inhibitor PD098059, indicating that the Raf-MEK-MAPK cascade is required for their expression. lysophosphatidic acid 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 9858557-8 1999 LPA-stimulated expression of c-Fos, Fra-1, Fra-2, c-Jun, and JunB is inhibited by the MEK1 inhibitor PD098059, indicating that the Raf-MEK-MAPK cascade is required for their expression. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 101-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 9858557-10 1999 The induction of c-Fos observed in response to LPA was strongly inhibited by buffering the intracellular [Ca2+]. lysophosphatidic acid 47-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 10051203-10 1999 Interestingly, the strong and widespread induction of both immediate-early gene transcripts was almost totally inhibited by para-chlorophenylalanine treatment; in the hypothalamic paraventricular nucleus for example, c-fos messenger RNA signal and the number of Fos-immunoreactive positive cells were reduced by 80 and 48%, respectively, in serotonin-depleted rats treated with the bacterial endotoxin. N-Chlorophenylalanine 129-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 217-222 11122538-1 1999 We have modified the cell-based directed cytotoxicity assay for sodium channel and calcium channel active phycotoxins using a c-fos-luciferase reporter gene construct. phycotoxins 106-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 11122538-6 1999 Saxitoxin caused a concentration-dependent inhibition of brevetoxin-1 induction of c-fos-luc with an EC50 of 3.5 ng ml(-1). Saxitoxin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 10391472-12 1999 dose of SKF 38393 (that produced rotation but not a net decrease in firing rate in basal ganglia output nuclei) induced Fos-like immunoreactivity in the striatum and subthalamic nucleus, suggesting an activation of both inhibitory and excitatory afferents to the substantia nigra and entopeduncular nucleus. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 8-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-123 10408598-6 1999 Administration of N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine, a selective noradrenergic neurotoxin, suppressed the basal expression of c-fos messenger RNA. DSP 4 18-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 10408598-7 1999 The response of c-fos to photo-stimulation was also significantly lower in the visual cortex of N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine-treated young rats. DSP 4 96-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 10408602-5 1999 Apomorphine and quinpirole increased the discharge rate and induced a strong expression of c-Fos-like immunoreactive proteins, whereas SKF 82958 induced a decrease of the discharge rate and a slight expression of c-Fos in 6-hydroxydopamine-lesioned rats. Apomorphine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 10408602-5 1999 Apomorphine and quinpirole increased the discharge rate and induced a strong expression of c-Fos-like immunoreactive proteins, whereas SKF 82958 induced a decrease of the discharge rate and a slight expression of c-Fos in 6-hydroxydopamine-lesioned rats. Apomorphine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-218 10408602-5 1999 Apomorphine and quinpirole increased the discharge rate and induced a strong expression of c-Fos-like immunoreactive proteins, whereas SKF 82958 induced a decrease of the discharge rate and a slight expression of c-Fos in 6-hydroxydopamine-lesioned rats. Quinpirole 16-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 10408602-5 1999 Apomorphine and quinpirole increased the discharge rate and induced a strong expression of c-Fos-like immunoreactive proteins, whereas SKF 82958 induced a decrease of the discharge rate and a slight expression of c-Fos in 6-hydroxydopamine-lesioned rats. SK and F 82958 135-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-218 10408602-5 1999 Apomorphine and quinpirole increased the discharge rate and induced a strong expression of c-Fos-like immunoreactive proteins, whereas SKF 82958 induced a decrease of the discharge rate and a slight expression of c-Fos in 6-hydroxydopamine-lesioned rats. Oxidopamine 222-239 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-218 10408610-0 1999 Central serotonin depletion modulates the behavioural, endocrine and physiological responses to repeated social stress and subsequent c-fos expression in the brains of male rats. Serotonin 8-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-139 10426492-7 1999 Compared to vehicle, glyceryl trinitrate-induced hypotension caused a marked induction of Fos protein in the caudal one-third of the nucleus tractus solitarius (bregma -14 to -13.3 mm), which tailed off rapidly in more rostral sections. Nitroglycerin 21-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 10426498-7 1999 Our results show that recurrent flurothyl-induced seizures progressively increased excitability of the brain, as revealed by a dramatic increase in the extent and intensity of c-fos immunostaining. Flurothyl 32-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 176-181 10426541-0 1999 The distribution of 3,4-methylenedioxymethamphetamine "Ecstasy"-induced c-fos expression in rat brain. N-Methyl-3,4-methylenedioxyamphetamine 20-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 10426541-0 1999 The distribution of 3,4-methylenedioxymethamphetamine "Ecstasy"-induced c-fos expression in rat brain. N-Methyl-3,4-methylenedioxyamphetamine 55-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 10426541-3 1999 Dose-dependent increases in c-fos expression were seen in much of the cortex, forebrain, brainstem and cerebellum in rats given 3,4-methylenedioxymethamphetamine. N-Methyl-3,4-methylenedioxyamphetamine 128-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 10426541-9 1999 Robust c-fos expression was seen in the cerebellum, particularly in the flocculus, and this may explain the reported deleterious effects of 3,4-methylenedioxymethamphetamine on balance and co-ordination. N-Methyl-3,4-methylenedioxyamphetamine 140-173 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 7-12 10426541-10 1999 Significant c-fos expression was also seen in the ventral tegmental area, amidst the cell bodies of mesolimbic and mesocortical dopamine neurons, and in the median and dorsal raphe, where the serotonergic innervation of the forebrain originates. Dopamine 128-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 10426541-11 1999 Double-labelling of fos-positive neurons with 5-hydroxytryptamine showed that only a small number of serotonergic neurons in the raphe expressed c-fos following 3,4-methylenedioxymethamphetamine. N-Methyl-3,4-methylenedioxyamphetamine 161-194 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-150 10426541-12 1999 The widespread distribution of 3,4-methylenedioxymethamphetamine-induced c-fos expression seen in this study can be linked to the profound alterations in physiological function, mood and behaviour produced by this drug. N-Methyl-3,4-methylenedioxyamphetamine 31-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 12840877-0 1999 Studies on hepatocyte apoptosis, proliferation and oncogene c-fos expression in carbon tetrachloride-induced cirrhotic rat liver. Carbon Tetrachloride 80-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 10336130-0 1999 Phosphorylation of transcription factor cyclic-AMP response element binding protein mediates c-fos induction elicited by sustained hypertension in rat nucleus tractus solitarii. Cyclic AMP 40-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 10336130-1 1999 We investigated the role of cyclic-AMP response element binding protein signaling in the induction of the immediate-early gene c-fos by baroreceptor activation in neurons of the nucleus tractus solitarii of anesthetized rats. Cyclic AMP 28-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 10336130-3 1999 This implied increase in phosphorylation of cyclic-AMP response element binding protein was subsequently followed by an elevation in the expression of Fos protein in neurons of the nucleus tractus solitarii. Cyclic AMP 44-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-154 10336130-7 1999 These findings suggest that phosphorylation of cyclic-AMP response element binding protein is crucial to Fos expression in the nucleus tractus solitarii elicited by sustained hypertension. Cyclic AMP 47-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-108 10336130-8 1999 As such, phosphorylation of cyclic-AMP response element binding protein may be an important early nuclear event that mediates the long-term inhibitory modulation of the baroreceptor reflex response by Fos protein at the nucleus tractus solitarii. Cyclic AMP 28-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 201-204 10366018-0 1999 Blockade of cannabinoid receptors by SR141716 selectively increases Fos expression in rat mesocorticolimbic areas via reduced dopamine D2 function. Rimonabant 37-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 10366018-2 1999 Intraperitoneal administration of the selective CB1 receptor antagonist, SR141716, dose-dependently (1.0, 3.0 and 10 mg/kg) increased Fos-like immunoreactivity in mesocorticolimbic areas (prefrontal cortex, ventrolateral septum, shell of the nucleus accumbens and dorsomedial caudate-putamen), while motor-related structures such as the core of the nucleus accumbens and the dorsolateral caudate-putamen were unaffected. Rimonabant 73-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-137 10051203-10 1999 Interestingly, the strong and widespread induction of both immediate-early gene transcripts was almost totally inhibited by para-chlorophenylalanine treatment; in the hypothalamic paraventricular nucleus for example, c-fos messenger RNA signal and the number of Fos-immunoreactive positive cells were reduced by 80 and 48%, respectively, in serotonin-depleted rats treated with the bacterial endotoxin. N-Chlorophenylalanine 129-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 262-265 10501478-0 1999 Sexual dimorphism in the response to N-methyl-D-aspartate receptor antagonists and morphine on behavior and c-Fos induction in the rat brain. Morphine 83-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 10501478-15 1999 In the caudate-putamen, morphine-induced c-Fos expression was significantly reduced by NPC-17742 (30 min before morphine) in males and completely blocked in females. Morphine 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 10613497-7 1999 Cellular expression of the protein product of the immediate early gene, c-fos, was dramatically increased in the nucleus accumbens shell after ventral hippocampal N-methyl-D-aspartate infusion, and haloperidol, SCH-23390 and reserpine attenuated this effect. N-Methylaspartate 163-183 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 10613497-7 1999 Cellular expression of the protein product of the immediate early gene, c-fos, was dramatically increased in the nucleus accumbens shell after ventral hippocampal N-methyl-D-aspartate infusion, and haloperidol, SCH-23390 and reserpine attenuated this effect. Haloperidol 198-209 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 10613497-7 1999 Cellular expression of the protein product of the immediate early gene, c-fos, was dramatically increased in the nucleus accumbens shell after ventral hippocampal N-methyl-D-aspartate infusion, and haloperidol, SCH-23390 and reserpine attenuated this effect. SCH 23390 211-220 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 10613497-7 1999 Cellular expression of the protein product of the immediate early gene, c-fos, was dramatically increased in the nucleus accumbens shell after ventral hippocampal N-methyl-D-aspartate infusion, and haloperidol, SCH-23390 and reserpine attenuated this effect. Reserpine 225-234 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 10613502-9 1999 Moreover, when double-label immunocytochemistry was used to identify cFos- and catecholamine-positive neurons in the brainstem, it was found that catecholamine-positive neurons in the ventrolateral medulla and locus coeruleus showed a significant increase in cFos expression in response to suckling compared with non-resuckled and pup-exposure groups. Catecholamines 146-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-73 10613502-9 1999 Moreover, when double-label immunocytochemistry was used to identify cFos- and catecholamine-positive neurons in the brainstem, it was found that catecholamine-positive neurons in the ventrolateral medulla and locus coeruleus showed a significant increase in cFos expression in response to suckling compared with non-resuckled and pup-exposure groups. Catecholamines 146-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 259-263 10501478-2 1999 Previous studies have shown that, upon administration of morphine, the immediate early gene c-Fos is induced in the striatum, nucleus accumbens and cortex of the rat brain. Morphine 57-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 10501478-3 1999 This induction of c-Fos is reduced by administration of the N-methyl-D-aspartate receptor antagonist dizocilpine maleate. Dizocilpine Maleate 101-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 10501478-5 1999 In male rats treated with morphine (10 mg/kg, s.c.) and killed 2 h later, there was an induction of c-Fos in the dorsomedial caudate-putamen, the nucleus accumbens and in the intralaminar nuclei of the thalamus. Morphine 26-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 10501478-8 1999 In females, morphine induced c-Fos in the caudate-putamen, but with more inter-animal variability than in males. Morphine 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 10501478-10 1999 In male rats, dizocilpine maleate alone caused negligible induction of c-Fos, whereas in female rats, it caused a large induction in the rhomboid, reuniens and central medial nuclei of the thalamus, and in the cortex. Dizocilpine Maleate 14-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 10501478-11 1999 Whereas dizocilpine maleate partially blocked the morphine-induced c-Fos expression in the caudate-putamen of males, it completely blocked this response in females. Dizocilpine Maleate 8-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 10501478-11 1999 Whereas dizocilpine maleate partially blocked the morphine-induced c-Fos expression in the caudate-putamen of males, it completely blocked this response in females. Morphine 50-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 10501478-15 1999 In the caudate-putamen, morphine-induced c-Fos expression was significantly reduced by NPC-17742 (30 min before morphine) in males and completely blocked in females. Morphine 112-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 10366018-3 1999 In the ventrolateral septum, taken as a representative structure, the Fos-inducing effect of SR141716 (10 mg/kg) was maximal 2 h after injection and returned to near control levels by 4 h. Within the prefrontal cortex, SR141716 increased the number of Fos-positive cells predominantly in the infralimbic and prelimbic cortices, presumptive pyramidal cells being the major cell types in which Fos was induced. Rimonabant 93-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 10579212-0 1999 Prior D1 dopamine receptor stimulation is required to prime D2-mediated striatal Fos expression in 6-hydroxydopamine-lesioned rats. Oxidopamine 99-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-84 10366018-3 1999 In the ventrolateral septum, taken as a representative structure, the Fos-inducing effect of SR141716 (10 mg/kg) was maximal 2 h after injection and returned to near control levels by 4 h. Within the prefrontal cortex, SR141716 increased the number of Fos-positive cells predominantly in the infralimbic and prelimbic cortices, presumptive pyramidal cells being the major cell types in which Fos was induced. Rimonabant 93-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 252-255 10366018-3 1999 In the ventrolateral septum, taken as a representative structure, the Fos-inducing effect of SR141716 (10 mg/kg) was maximal 2 h after injection and returned to near control levels by 4 h. Within the prefrontal cortex, SR141716 increased the number of Fos-positive cells predominantly in the infralimbic and prelimbic cortices, presumptive pyramidal cells being the major cell types in which Fos was induced. Rimonabant 93-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 252-255 10366018-3 1999 In the ventrolateral septum, taken as a representative structure, the Fos-inducing effect of SR141716 (10 mg/kg) was maximal 2 h after injection and returned to near control levels by 4 h. Within the prefrontal cortex, SR141716 increased the number of Fos-positive cells predominantly in the infralimbic and prelimbic cortices, presumptive pyramidal cells being the major cell types in which Fos was induced. Rimonabant 219-227 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 10579212-3 1999 Our prior work has shown that three pretreatment injections of the mixed D1/D2 agonist apomorphine (0.5 mg/kg) into 6-hydroxydopamine-lesioned rats permits a previously inactive dose of the D2 agonist quinpirole (0.25 mg/kg) to induce robust contralateral rotation and striatal Fos expression in striatoentopeduncular "direct" pathway neurons. Apomorphine 87-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 278-281 10366018-5 1999 By contrast, the dopamine D2-like agonist, quinpirole (0.25 mg/ kg), counteracted SR141716-induced Fos-like immunoreactivity in the ventrolateral septum, the nucleus accumbens and the dorsomedial caudate-putamen, while no antagonism was observed in the prefrontal cortex. Dopamine 17-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 10604276-3 1999 In in situ hybridization using [35S]UTP-labeled antisense c-fos, cRNA increased c-fos mRNA levels through the hippocampal formation, piriform cortex, septum, caudate-putamen, neostriatum, and amygdala within 2 hr. Uridine Triphosphate 36-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 10366018-5 1999 By contrast, the dopamine D2-like agonist, quinpirole (0.25 mg/ kg), counteracted SR141716-induced Fos-like immunoreactivity in the ventrolateral septum, the nucleus accumbens and the dorsomedial caudate-putamen, while no antagonism was observed in the prefrontal cortex. Quinpirole 43-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 10366018-5 1999 By contrast, the dopamine D2-like agonist, quinpirole (0.25 mg/ kg), counteracted SR141716-induced Fos-like immunoreactivity in the ventrolateral septum, the nucleus accumbens and the dorsomedial caudate-putamen, while no antagonism was observed in the prefrontal cortex. Rimonabant 82-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 10366018-6 1999 Microdialysis experiments in awake rats indicated that SR141716, at doses which increased Fos expression (3 and 10 mg/kg), did not alter dopamine release in the shell of the nucleus accumbens. Rimonabant 55-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 10391434-0 1999 The delayed effects of phencyclidine enhance amphetamine-induced behavior and striatal C-Fos expression in the rat. Phencyclidine 23-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 10391434-5 1999 Likewise, phencyclidine pretreatment produced an increase in the number of striatal cells expressing c-Fos following treatment with 0.5 mg/kg amphetamine. Phencyclidine 10-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 10391434-5 1999 Likewise, phencyclidine pretreatment produced an increase in the number of striatal cells expressing c-Fos following treatment with 0.5 mg/kg amphetamine. Amphetamine 142-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 10391434-6 1999 Phencyclidine pretreatment did not alter c-Fos induction in the nucleus accumbens, but did decrease the basal number of c-Fos-containing cells in the anterior cingulate cortex. Phencyclidine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 10391434-8 1999 These data demonstrate that the delayed effects of a single dose of phencyclidine alter anterior cingulate cortex neurochemistry, and enhance the behavioral and striatal c-Fos response to a low dose of amphetamine. Phencyclidine 68-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-175 10391434-8 1999 These data demonstrate that the delayed effects of a single dose of phencyclidine alter anterior cingulate cortex neurochemistry, and enhance the behavioral and striatal c-Fos response to a low dose of amphetamine. Amphetamine 202-213 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-175 10604276-6 1999 The present study indicates that lidocaine induces convulsions and c-fos expression without causing neurotoxicity. Lidocaine 33-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 9881578-0 1998 The effect of E-5842, a sigma receptor ligand and potential atypical antipsychotic, on Fos expression in rat forebrain. E 5842 14-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 9881578-1 1998 We examined the ability of E-5842 (4-(4-fluorophenil)-1,2,3,6- tetrahydro-1-[4-(1,2,4-triazol-1-il)butyl] pyridine citrate), a sigma receptor ligand, to increase Fos protein expression in regions of rat forebrain. E 5842 27-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-165 9881578-1 1998 We examined the ability of E-5842 (4-(4-fluorophenil)-1,2,3,6- tetrahydro-1-[4-(1,2,4-triazol-1-il)butyl] pyridine citrate), a sigma receptor ligand, to increase Fos protein expression in regions of rat forebrain. 4-(4-fluorophenil)-1,2,3,6- tetrahydro-1-[4-(1,2,4-triazol-1-il)butyl] pyridine citrate 35-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-165 9838105-1 1998 Previous studies from this laboratory have demonstrated that acute administration of morphine induces the immediate-early genes (IEGs) c-Fos and JunB in the rat caudate putamen (CPu). Morphine 85-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 10604276-0 1999 Systemic injection of lidocaine induced expression of c-fos mRNA and protein in adult rat brain. Lidocaine 22-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 10604276-2 1999 In this study, we examined whether the intraperitoneal injections of lidocaine at doses inducing convulsions within 10 min, increased the level of c-fos mRNA and protein in forebrain areas. Lidocaine 69-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-152 10604276-3 1999 In in situ hybridization using [35S]UTP-labeled antisense c-fos, cRNA increased c-fos mRNA levels through the hippocampal formation, piriform cortex, septum, caudate-putamen, neostriatum, and amygdala within 2 hr. Sulfur-35 32-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 10604276-3 1999 In in situ hybridization using [35S]UTP-labeled antisense c-fos, cRNA increased c-fos mRNA levels through the hippocampal formation, piriform cortex, septum, caudate-putamen, neostriatum, and amygdala within 2 hr. Sulfur-35 32-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 10604276-3 1999 In in situ hybridization using [35S]UTP-labeled antisense c-fos, cRNA increased c-fos mRNA levels through the hippocampal formation, piriform cortex, septum, caudate-putamen, neostriatum, and amygdala within 2 hr. Uridine Triphosphate 36-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 9838105-5 1998 Morphine significantly induced c-Fos expression in the dorsomedial CPu, as we have reported previously. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 9838105-6 1998 Whereas MDL 72222 alone did not induce c-Fos, it potentiated the morphine-induced c-Fos expression. Morphine 65-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 9838135-6 1998 Results show that kainic acid induced c-fos synthesis in most of these glial cells. Kainic Acid 18-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 9928001-4 1998 The effects of PACAP on c-fos gene expression and granule cell survival were both mimicked by dbcAMP but not by PMA. Bucladesine 94-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 9852594-3 1998 We found, using in situ hybridization of c-fos mRNA, that the pattern of neuronal activation in the cortex, in the caudate, in the shell and core of the nucleus accumbens, and in other subcortical structures was markedly different when amphetamine (2.0 mg/kg, i.p.) Amphetamine 236-247 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 9852594-5 1998 In most brain regions the magnitude of c-fos expression was over two times greater in rats given amphetamine plus novelty than in rats given amphetamine alone. Amphetamine 97-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 9852594-5 1998 In most brain regions the magnitude of c-fos expression was over two times greater in rats given amphetamine plus novelty than in rats given amphetamine alone. Amphetamine 141-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 9852594-7 1998 Furthermore, a unilateral 6-hydroxydopamine lesion of the mesostriatal dopamine system reduced amphetamine- but not novelty-induced c-fos expression. Oxidopamine 26-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 9852594-7 1998 Furthermore, a unilateral 6-hydroxydopamine lesion of the mesostriatal dopamine system reduced amphetamine- but not novelty-induced c-fos expression. Dopamine 35-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 9838035-0 1998 Intrastriatal and intraventricular injections of oligodeoxynucleotides in the rat brain: tissue penetration, intracellular distribution and c-fos antisense effects. Oligodeoxyribonucleotides 49-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 9838035-2 1998 These time and space parameters were correlated with the ability of c-fos phosphorothioate antisense ODNs to suppress the induction of Fos protein by cocaine. Parathion 74-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 9838035-2 1998 These time and space parameters were correlated with the ability of c-fos phosphorothioate antisense ODNs to suppress the induction of Fos protein by cocaine. Parathion 74-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-138 9838035-2 1998 These time and space parameters were correlated with the ability of c-fos phosphorothioate antisense ODNs to suppress the induction of Fos protein by cocaine. Cocaine 150-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 9838035-2 1998 These time and space parameters were correlated with the ability of c-fos phosphorothioate antisense ODNs to suppress the induction of Fos protein by cocaine. Cocaine 150-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-138 9838083-8 1998 Thus the immediate cell activation induced by lithium-pilocarpine seizures which is present at all ages translates only into a late wave of c-Fos and the expression of HSP72 in P21 and adult animals in which there will be extensive cell damage. Lithium 46-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 9838083-8 1998 Thus the immediate cell activation induced by lithium-pilocarpine seizures which is present at all ages translates only into a late wave of c-Fos and the expression of HSP72 in P21 and adult animals in which there will be extensive cell damage. Pilocarpine 54-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 9874176-6 1998 Phorbol myristate acetate was also able to increase c-fos mRNA expression. Tetradecanoylphorbol Acetate 0-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 9886869-7 1998 More recent experiments in the spontaneously hypertensive rat (SHR) using the immediate-early gene c-fos as a marker of neuronal activity, have demonstrated that impaired activity of this short inhibitory GABA pathway in the SHR disinhibits the bulbospinal pressor pathway, thus contributing to the hypertension in this model. gamma-Aminobutyric Acid 205-209 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 9879660-2 1998 Transcript levels for both c-fos and glyceraldehyde-3 -phosphate dehydrogenase were determined by Northern blot analysis using 32P-labeled cDNA probes. Phosphorus-32 127-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 9879660-4 1998 Treatment of PC12 cells with a combination of agents (NGF, forskolin, and tetradecanoylphorbol acetate [TPA]) increased c-fos expression over that detected with NGF alone. Colforsin 59-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 9879660-4 1998 Treatment of PC12 cells with a combination of agents (NGF, forskolin, and tetradecanoylphorbol acetate [TPA]) increased c-fos expression over that detected with NGF alone. Tetradecanoylphorbol Acetate 74-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 9879660-4 1998 Treatment of PC12 cells with a combination of agents (NGF, forskolin, and tetradecanoylphorbol acetate [TPA]) increased c-fos expression over that detected with NGF alone. Tetradecanoylphorbol Acetate 104-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 9879660-7 1998 It was determined that MF exposure, like superinduction with anisomycin, increased c-fos expression only in cultures which were not yet exhibiting maximal c-fos expression. Anisomycin 61-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 9759976-12 1998 Additionally, examination of FOS-like immunoreactivity revealed a distinct pattern following L-(+)-2-amino-4-phosphonobutyrate administration in 6-hydroxydopamine lesioned versus intact rats. 2-amino-4-phosphonobutyric acid 93-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 9759976-12 1998 Additionally, examination of FOS-like immunoreactivity revealed a distinct pattern following L-(+)-2-amino-4-phosphonobutyrate administration in 6-hydroxydopamine lesioned versus intact rats. Oxidopamine 145-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 9870571-5 1998 Large amounts of LPS however increased the number of capsaicin-induced c-fos positive cells in the TNC I,II. Capsaicin 53-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 9824685-0 1998 Development of tolerance to the antinociceptive effect of systemic morphine at the lumbar spinal cord level: a c-Fos study in the rat. Morphine 67-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 9824685-1 1998 The development of tolerance to the antinociceptive effects of morphine was investigated in rats using carrageenin-induced spinal c-Fos expression. Morphine 63-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 9824685-1 1998 The development of tolerance to the antinociceptive effects of morphine was investigated in rats using carrageenin-induced spinal c-Fos expression. Carrageenan 103-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 9824685-3 1998 Two hours after intraplantar injection of carrageenin (6 mg/150 microliter of saline), c-Fos-like immunoreactivity (FLI) was observed predominantly in the superficial and deep laminae of the dorsal horn in segments L4 and L5 of the spinal cord. Carrageenan 42-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 9824685-3 1998 Two hours after intraplantar injection of carrageenin (6 mg/150 microliter of saline), c-Fos-like immunoreactivity (FLI) was observed predominantly in the superficial and deep laminae of the dorsal horn in segments L4 and L5 of the spinal cord. Sodium Chloride 78-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 9857953-2 1998 The presence of DNIC was revealed by the reduction in the levels of Fos-like immunoreactivity that are normally induced by a standardized primary pinch stimulus when this stimulus was accompanied by a concurrent noxious stimulus (formalin) to a heterotopic body part. Formaldehyde 230-238 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 9802702-0 1998 Immunohistochemical localization of caffeine-induced c-Fos protein expression in the rat brain. Caffeine 36-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 9802702-8 1998 In contrast, caffeine at 75 mg/kg induced a significant increase compared with the saline condition in the number of Fos-immunoreactive neurons in the majority of structures examined. Caffeine 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 9802702-8 1998 In contrast, caffeine at 75 mg/kg induced a significant increase compared with the saline condition in the number of Fos-immunoreactive neurons in the majority of structures examined. Sodium Chloride 83-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 9811905-9 1998 Analysis of the mean number of c-fos immunoreactive cell nuclei showed a significant reduction in the estradiol benzoate versus control groups in areas of the forebrain relating to sensory, contextual, and integrative processing. estradiol 3-benzoate 102-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 9804869-3 1998 Within the first hour after a single dose of dexamethasone, and intensifying over 4 h, marked induction of brain c-fos was seen. Dexamethasone 45-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 9890606-0 1999 Persistent c-fos induction by nicotine in developing rat brain regions: interaction with hypoxia. Nicotine 30-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 9890606-2 1999 We administered nicotine to pregnant rats throughout gestation and neonatal brains were examined for expression of c-fos, a nuclear transcription factor involved in differentiation and cell death. Nicotine 16-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 9890606-3 1999 The nicotine group showed persistent c-fos overexpression in the forebrain long after termination of exposure; in the brainstem, overexpression was apparent both after birth and at the end of the second postnatal week. Nicotine 4-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 9890606-6 1999 Hypoxia evoked acute stimulation of c-fos with a regional selectivity and ontogenetic profile differing from those of prenatal nicotine and this acute response was reduced by prenatal nicotine treatment. Nicotine 184-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 9890606-7 1999 Persistent c-fos elevation is a harbinger of cell death, a relationship that provides an underlying mechanism for eventual cell deficits that appear after fetal nicotine exposure. Nicotine 161-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 9890606-8 1999 Nicotine"s interference with the acute c-fos stimulation caused by a subsequent episode of hypoxia may indicate a further compromise of cellular repair mechanisms. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 9843881-0 1998 The area postrema modulates hypothalamic fos responses to intragastric hypertonic saline in conscious rats. Sodium Chloride 82-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 9843881-1 1998 We have recently reported that an acute intragastric hypertonic saline load increases Fos immunoreactivity in several central nuclei, including the supraoptic nucleus (SON), paraventricular nucleus (PVN), nucleus of the solitary tract (NTS), area postrema (AP), and lateral parabrachial nucleus (LPBN). Sodium Chloride 64-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 9842840-0 1998 Propofol inhibits ketamine-induced c-fos expression in the rat posterior cingulate cortex. Propofol 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 9842840-0 1998 Propofol inhibits ketamine-induced c-fos expression in the rat posterior cingulate cortex. Ketamine 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 9842840-4 1998 We investigated the effect of propofol, which has both GABAA receptor-activating and NMDA receptor-suppressing activity, on ketamine-induced c-fos expression in the rat posterior cingulate cortex. Propofol 30-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 9842840-4 1998 We investigated the effect of propofol, which has both GABAA receptor-activating and NMDA receptor-suppressing activity, on ketamine-induced c-fos expression in the rat posterior cingulate cortex. Ketamine 124-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 9842840-8 1998 Propofol significantly inhibited ketamine-induced c-fos expression in the posterior cingulate cortex. Propofol 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 9842840-8 1998 Propofol significantly inhibited ketamine-induced c-fos expression in the posterior cingulate cortex. Ketamine 33-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 9842840-11 1998 IMPLICATIONS: In the present study, we demonstrated that the clinically relevant dose of propofol significantly inhibited ketamine-induced c-fos expression in the rat posterior cingulate cortex. Propofol 89-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 9842840-11 1998 IMPLICATIONS: In the present study, we demonstrated that the clinically relevant dose of propofol significantly inhibited ketamine-induced c-fos expression in the rat posterior cingulate cortex. Ketamine 122-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 9879931-0 1998 Induction of c-fos, and cytochrome c oxidase subunits I and II by gossypol acetic acid in rat liver cells. gossypol acetic acid 66-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 9879931-2 1998 In northern blot analysis of gossypol-treated cells, there was a dose-dependent increase in c-fos, a component of the redox-regulated transcription factor activator protein-1 (AP-1). Gossypol 29-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 9879931-4 1998 A rapid 3.33+/-1.37 fold induction in c-fos was detected after treatment with 5 micromol/L gossypol for 30 min. Gossypol 91-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 9879931-5 1998 Additionally, treatment with 5 micromol/L gossypol for 12 h caused a 2.66+/-0.67 fold increase in c-fos expression. Gossypol 42-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 9824467-0 1998 Modification of haloperidol-induced pattern of c-fos expression by serotonin agonists. Haloperidol 16-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 9824467-1 1998 Acute challenge with clozapine and haloperidol produce different anatomical patterns of c-fos expression in the forebrain. Clozapine 21-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 9824467-1 1998 Acute challenge with clozapine and haloperidol produce different anatomical patterns of c-fos expression in the forebrain. Haloperidol 35-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 9824467-3 1998 In order to test this possibility, we examined the abilities of 5-HT1A and 5-HT2A/2c agonists to modify the pattern of c-fos expression induced by haloperidol and clozapine. Haloperidol 147-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 9824467-3 1998 In order to test this possibility, we examined the abilities of 5-HT1A and 5-HT2A/2c agonists to modify the pattern of c-fos expression induced by haloperidol and clozapine. Clozapine 163-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 9824467-6 1998 In saline-pretreated rats, haloperidol produced intense Fos-LI in all four striatal quadrants while the effect of clozapine was restricted to the medial part of the striatum. Sodium Chloride 3-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 9824467-6 1998 In saline-pretreated rats, haloperidol produced intense Fos-LI in all four striatal quadrants while the effect of clozapine was restricted to the medial part of the striatum. Haloperidol 27-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 9824467-7 1998 Prior administration of 8-OH-DPAT significantly reduced haloperidol-induced Fos-LI in all four striatal quadrants while DOI and 8-OHDPAT + DOI significantly reduced Fos-LI only in dorso- and ventrolateral quadrants. 8-Hydroxy-2-(di-n-propylamino)tetralin 24-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 9824467-7 1998 Prior administration of 8-OH-DPAT significantly reduced haloperidol-induced Fos-LI in all four striatal quadrants while DOI and 8-OHDPAT + DOI significantly reduced Fos-LI only in dorso- and ventrolateral quadrants. Haloperidol 56-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 9824467-7 1998 Prior administration of 8-OH-DPAT significantly reduced haloperidol-induced Fos-LI in all four striatal quadrants while DOI and 8-OHDPAT + DOI significantly reduced Fos-LI only in dorso- and ventrolateral quadrants. 8-Hydroxy-2-(di-n-propylamino)tetralin 128-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-168 9824467-8 1998 In the nucleus accumbens, haloperidol induced intense Fos-LI in the core and the shell regions whereas clozapine induced c-fos expression only in the shell. Haloperidol 26-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 9824467-8 1998 In the nucleus accumbens, haloperidol induced intense Fos-LI in the core and the shell regions whereas clozapine induced c-fos expression only in the shell. Clozapine 103-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-126 9824467-9 1998 Pretreatment with 8-OHDPAT in haloperidol treated rats reduced Fos-LI in the core region yielding to a c-fos pattern similar to that induced by clozapine. 8-Hydroxy-2-(di-n-propylamino)tetralin 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 9824467-9 1998 Pretreatment with 8-OHDPAT in haloperidol treated rats reduced Fos-LI in the core region yielding to a c-fos pattern similar to that induced by clozapine. 8-Hydroxy-2-(di-n-propylamino)tetralin 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 9824467-9 1998 Pretreatment with 8-OHDPAT in haloperidol treated rats reduced Fos-LI in the core region yielding to a c-fos pattern similar to that induced by clozapine. Haloperidol 30-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 9824467-9 1998 Pretreatment with 8-OHDPAT in haloperidol treated rats reduced Fos-LI in the core region yielding to a c-fos pattern similar to that induced by clozapine. Haloperidol 30-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 9824467-10 1998 In the prefrontal cortex of saline-pretreated rats, haloperidol produced a moderate c-fos expression compared with the intense expression produced by clozapine. Sodium Chloride 28-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 9824467-10 1998 In the prefrontal cortex of saline-pretreated rats, haloperidol produced a moderate c-fos expression compared with the intense expression produced by clozapine. Haloperidol 52-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 9886788-1 1998 We used immunohistochemical detection of the Fos protein to study the neuronal activation in the brain of methoxyfluorane-anesthetized rats after noxious deep somatic or visceral stimulation. methoxyfluorane 106-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 9824467-12 1998 These results show the ability of 5-HT agonists to transform the typical pattern of c-fos expression induced by haloperidol into a pattern resembling that of clozapine. Haloperidol 112-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 9822457-2 1998 Microinjection into the bilateral nucleus tractus solitarii of an antisense oligonucleotide that targets against the initiation codon of c-fos mRNA significantly potentiated the baroreceptor reflex in response to 30 minutes of sustained increase in blood pressure. Oligonucleotides 76-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 9822457-5 1998 Control treatment with the corresponding sense oligonucleotide, an antisense oligonucleotide that targets against a different portion of the coding sequence of the c-fos mRNA or artificial cerebrospinal fluid, on the other hand, elicited no discernible effect on either the baroreceptor reflex response or the induced expression of Fos protein in the nucleus tractus solitarii by baroreceptor activation. Oligonucleotides 47-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-169 9822457-5 1998 Control treatment with the corresponding sense oligonucleotide, an antisense oligonucleotide that targets against a different portion of the coding sequence of the c-fos mRNA or artificial cerebrospinal fluid, on the other hand, elicited no discernible effect on either the baroreceptor reflex response or the induced expression of Fos protein in the nucleus tractus solitarii by baroreceptor activation. Oligonucleotides 77-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-169 9831260-7 1998 Phenylephrine infusion and CCK injection elicited Fos expression in distinct and in overlapping regions of the NTS and the VLM. Phenylephrine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 9831260-10 1998 In contrast, PE infusion produced a more restricted distribution of Fos-positive neurones in the NTS, with most neurones confined to a dorsolateral strip containing few TH-positive neurones. Phenylephrine 13-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 9831261-7 1998 Experiments on c-fos gene expression in these cultures showed that while only 12% of the magnocellular OT and VP neurones contained barely detectable Fos protein in their nuclei under control conditions, potassium depolarization of these cultures for 3 h produced intense c-fos expression in 87-91% of these cells. Potassium 204-213 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 272-277 9795122-7 1998 In addition to the behavioral sensitization, Amp-pretreated rats showed a reduction in the ability of the acute Amp challenge to induce c-fos mRNA in the medial prefrontal cortex and neurotensin/neuromedin N (NT/N) mRNA in the nucleus accumbens-shell. Amphetamine 45-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 9795122-7 1998 In addition to the behavioral sensitization, Amp-pretreated rats showed a reduction in the ability of the acute Amp challenge to induce c-fos mRNA in the medial prefrontal cortex and neurotensin/neuromedin N (NT/N) mRNA in the nucleus accumbens-shell. Amphetamine 112-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 9795122-8 1998 At doses that blocked the initiation of behavioral sensitization to Amp, clozapine fully and haloperidol partially restored the capacity of acute Amp to induce c-fos and NT/N gene expression. Haloperidol 93-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 9795122-8 1998 At doses that blocked the initiation of behavioral sensitization to Amp, clozapine fully and haloperidol partially restored the capacity of acute Amp to induce c-fos and NT/N gene expression. Amphetamine 146-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 9795131-1 1998 The induction of c-fos mRNA in rat brain due to morphine treatment was analyzed by in situ hybridization. Morphine 48-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 9795131-3 1998 However, rats that were repeatedly pretreated with morphine displayed a marked c-fos induction in a few brain areas in response to morphine application. Morphine 51-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 9795131-3 1998 However, rats that were repeatedly pretreated with morphine displayed a marked c-fos induction in a few brain areas in response to morphine application. Morphine 131-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 9795131-6 1998 Naloxone-precipitated withdrawal led to a more intense c-fos expression which also encompassed a greater range of brain areas. Naloxone 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 9795131-8 1998 A low morphine dose suppressed the c-fos expression nearly completely and was not sufficient to elicit the morphine-like expression pattern of c-fos. Morphine 6-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 9795131-11 1998 These areas responded to morphine with an elevated c-fos expression only when morphine was repeatedly given previously. Morphine 25-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 9795131-11 1998 These areas responded to morphine with an elevated c-fos expression only when morphine was repeatedly given previously. Morphine 78-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 10437133-0 1998 NMDA receptors mediating Fos expression in rat spinal cord induced by subcutaneous injection of formalin. Formaldehyde 96-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 10437133-1 1998 AIM: To examine the effects of N-methyl-D-aspartate (NMDA) and non-NMDA receptors on noxious stimulation-induced Fos expression in the rat spinal cord. N-Methylaspartate 31-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-116 10437133-1 1998 AIM: To examine the effects of N-methyl-D-aspartate (NMDA) and non-NMDA receptors on noxious stimulation-induced Fos expression in the rat spinal cord. N-Methylaspartate 53-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-116 10437133-4 1998 RESULTS: Two hours after s.c. formalin, Fos-like immunoreactive (FLI) neurons were distributed mainly in medial part of the lamina I and the outer lamina II of the ipsilateral dorsal horn. Formaldehyde 30-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 9795171-0 1998 Methamphetamine induces fos expression in the striatum and the substantia nigra pars reticulata in a rat model of Parkinson"s disease. Methamphetamine 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 9795171-9 1998 It is suggested that the FLIs in the two discrete sites are activated independently by different mechanisms, and furthermore, different neuronal pathways are involved in the methamphetamine-induced rotation and Fos expression in the SNr of 6-OHDA rats. Methamphetamine 174-189 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 211-214 9795171-9 1998 It is suggested that the FLIs in the two discrete sites are activated independently by different mechanisms, and furthermore, different neuronal pathways are involved in the methamphetamine-induced rotation and Fos expression in the SNr of 6-OHDA rats. Oxidopamine 240-246 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 211-214 9855286-4 1998 Striatal c-Fos protein levels and AP-1 DNA binding activity were increased in rats receiving paired morphine compared with rats that did not receive morphine but not in rats receiving morphine without the CS. Morphine 100-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 9855286-4 1998 Striatal c-Fos protein levels and AP-1 DNA binding activity were increased in rats receiving paired morphine compared with rats that did not receive morphine but not in rats receiving morphine without the CS. Morphine 149-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 9855286-4 1998 Striatal c-Fos protein levels and AP-1 DNA binding activity were increased in rats receiving paired morphine compared with rats that did not receive morphine but not in rats receiving morphine without the CS. Morphine 149-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 9855286-4 1998 Striatal c-Fos protein levels and AP-1 DNA binding activity were increased in rats receiving paired morphine compared with rats that did not receive morphine but not in rats receiving morphine without the CS. Cesium 205-207 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 9855303-3 1998 The dose of SKF 38393 used to induce contralateral rotation also caused an intense Fos protein immunoreactivity in the rat dorsolateral striatum on the lesioned site which was inhibited by PTAC indicating that the inhibition of rotation by PTAC was not due to non-specific peripheral side effects. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 12-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 9767180-18 1998 Here, 1 h intracarotid glucose infusions (4 mg/kg/min) selectively increased Fos-like immunoreactivity (FLIR) in the hypothalamic paraventricular, ventromedial, dorsomedial and arcuate nuclei of inbred DR but not DIO rats. Glucose 23-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-80 9756985-0 1998 Effects of ZD6169, a KATP channel opener, on bladder hyperactivity and spinal c-fos expression evoked by bladder irritation in rats. Zeneca ZD 6169 11-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 9855303-3 1998 The dose of SKF 38393 used to induce contralateral rotation also caused an intense Fos protein immunoreactivity in the rat dorsolateral striatum on the lesioned site which was inhibited by PTAC indicating that the inhibition of rotation by PTAC was not due to non-specific peripheral side effects. 6-(3-propylthio-1,2,5-thiadiazol-4-yl)-1-azabicyclo(3.2.1)octane 189-193 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 9776348-4 1998 Three hours after intraplantar injection of carrageenan (6 mg in 150 microl of saline) in awake rats, a peripheral oedema and numerous c-Fos protein-like immunoreactive (c-Fos-LI) neurons in L4 L5 segments were observed. Carrageenan 44-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 9757037-5 1998 In addition to the proENK mRNA level, PGE2 also increased c-Fos (approximately 4.3-fold), Fra-1 ( approximately 3.8 fold), and Fra-2 (approximately 8.2-fold) protein levels at 4 h after drug treatment. Dinoprostone 38-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 9792328-0 1998 Role of endogenous opiates in glucoprivic inhibition of the luteinizing hormone surge and fos expression by preoptic gonadotropin-releasing hormone neurones in ovariectomized steroid-primed female rats. Steroids 175-182 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 9792328-9 1998 Dual-label immunocytochemistry of tissue sections from the preoptic area and anterior hypothalamus of OVX, steroid-primed rats revealed nuclear Fos-immunoreactivity (-ir) in a subpopulation of gonadotropin-releasing hormone-(GnRH-)immunopositive neurones prior to maximal preovulatory LH release. Luteinizing Hormone 285-287 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-147 9792328-10 1998 Animals pretreated with 2DG i.c.v showed a significant decrease in mean numbers of GnRH neurones exhibiting Fos-ir, whereas coadministration of 2DG and NALT resulted in numbers of double-labelled neurones that were similar to those detected in the non-drug-treated controls. Deoxyglucose 24-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 10190151-10 1998 c-Fos-like immunoreactivity, an indicator of neuronal activation, was detected in the ipsilateral, but not in the contralateral frontoparietal cortex 2 hr after KCl application. Potassium Chloride 161-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 10190151-15 1998 These findings suggest that the inhibitory effects of lomerizine and flunarizine on the interval between the initiated and subsequent spontaneous SDs, the cortical hypoperfusion and expression of c-Fos-like immunoreactivity induced by SD are mediated via the effects of Ca2+ entry blockade, which may include an increase in cerebral blood flow and the prevention of excessive Ca2+ influx into brain cells. lomerizine 54-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 196-201 10190151-15 1998 These findings suggest that the inhibitory effects of lomerizine and flunarizine on the interval between the initiated and subsequent spontaneous SDs, the cortical hypoperfusion and expression of c-Fos-like immunoreactivity induced by SD are mediated via the effects of Ca2+ entry blockade, which may include an increase in cerebral blood flow and the prevention of excessive Ca2+ influx into brain cells. Flunarizine 69-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 196-201 9739136-8 1998 Systemic morphine suppressed c-Fos expression dose-dependently in both superficial and deep layers of dorsal horn and the latter region was much more sensitive to morphine than the former one. Morphine 9-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 9739136-8 1998 Systemic morphine suppressed c-Fos expression dose-dependently in both superficial and deep layers of dorsal horn and the latter region was much more sensitive to morphine than the former one. Morphine 163-171 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 9739147-1 1998 The serotonin agonist quipazine has been shown to cause phase shifts in melatonin and activity rhythms and to induce c-fos in the suprachiasmatic nucleus of rats. Serotonin 4-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 9739147-1 1998 The serotonin agonist quipazine has been shown to cause phase shifts in melatonin and activity rhythms and to induce c-fos in the suprachiasmatic nucleus of rats. Quipazine 22-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 9739147-9 1998 Ritanserin was more potent than ketanserin at inhibiting the DOI induced increase in c-Fos labelled cells in the SCN. Ritanserin 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 9739147-9 1998 Ritanserin was more potent than ketanserin at inhibiting the DOI induced increase in c-Fos labelled cells in the SCN. Ketanserin 32-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 9727025-7 1998 In addition to the role of c-Fos in regulating dopamine beta-hydroxylase gene expression in response to cAMP, a second pathway, involving Rap1/B-Raf is involved. Cyclic AMP 104-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 9776348-4 1998 Three hours after intraplantar injection of carrageenan (6 mg in 150 microl of saline) in awake rats, a peripheral oedema and numerous c-Fos protein-like immunoreactive (c-Fos-LI) neurons in L4 L5 segments were observed. Carrageenan 44-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-175 9776348-7 1998 Intravenous racemic-flurbiprofen (0.3, 3 and 9 mg kg(-1)) dose-relatedly reduced the carrageenan-evoked oedema and spinal c-Fos expression (r=0.64, r=0.88 and r=0.84 for paw diameter, ankle diameter and number of c-Fos-LI neurons; P<0.05. Flurbiprofen 20-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 9776348-7 1998 Intravenous racemic-flurbiprofen (0.3, 3 and 9 mg kg(-1)) dose-relatedly reduced the carrageenan-evoked oedema and spinal c-Fos expression (r=0.64, r=0.88 and r=0.84 for paw diameter, ankle diameter and number of c-Fos-LI neurons; P<0.05. Flurbiprofen 20-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-218 9776348-10 1998 Similar effects to those of intravenous racemic-flurbiprofen were obtained with intravenous S(+)-flurbiprofen (0.3, 3 and 9 mg kg(-1)) which dose-relatedly reduced the number of c-Fos-LI neurons (r=0.69, P<0.01) and diameters of paw and ankle (r=0.56 and r=0.52 respectively, P<0.05 for both). Flurbiprofen 92-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 178-183 9776348-16 1998 Intraplantar racemic-flurbiprofen (1, 10 and 30 microg) dose-relatedly reduced the carrageenan-enhanced ankle diameter (r=0.81, P<0.001) and the number of c-Fos-LI neurons in L4-L5 segments (r=0.83, P<0.001). Flurbiprofen 21-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 9776348-19 1998 For intraplantar S(+)-flurbiprofen (1, 10 and 30 microg) the dose-related effects (r=0.77, r=0.60 and r=0.59 for c-Fos-LI neurons, paw and ankle diameters respectively, P<0.001, P<0.01 and P<0.01) were similar to those of racemic-flurbiprofen. Flurbiprofen 18-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 9776348-19 1998 For intraplantar S(+)-flurbiprofen (1, 10 and 30 microg) the dose-related effects (r=0.77, r=0.60 and r=0.59 for c-Fos-LI neurons, paw and ankle diameters respectively, P<0.001, P<0.01 and P<0.01) were similar to those of racemic-flurbiprofen. Flurbiprofen 22-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 9737720-0 1998 Glucose and glucoincretin peptides synergize to induce c-fos, c-jun, junB, zif-268, and nur-77 gene expression in pancreatic beta(INS-1) cells. Glucose 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 9740405-0 1998 Fluoxetine induces the transcription of genes encoding c-fos, corticotropin-releasing factor and its type 1 receptor in rat brain. Fluoxetine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 9740405-2 1998 The present experiments were carried out to assess the effects of fluoxetine on c-fos induction throughout the rat brain. Fluoxetine 66-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 9721162-0 1998 Gastric acid-evoked c-fos messenger RNA expression in rat brainstem is signaled by capsaicin-resistant vagal afferents. Capsaicin 83-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 9721162-3 1998 METHODS: Neuronal excitation in the rat brainstem and spinal cord after intragastric administration of HCl (0.35-0.7 mol/L) was examined by in situ hybridization autoradiography for the immediate early gene c-fos. Hydrochloric Acid 103-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 207-212 9737720-7 1998 A role for Ca2+ signaling is inferred, since the L-type Ca2+ channel blocker nifedipine markedly reduces the induction of c-fos and nur-77 by glucose and GLP-1. Nifedipine 77-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 9737720-2 1998 Glucose causes a coordinated transcriptional activation of the IEGs c-fos, c-jun, JunB, zif-268, and nur-77 in the pancreatic beta-cell line INS-1. Glucose 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 9737720-7 1998 A role for Ca2+ signaling is inferred, since the L-type Ca2+ channel blocker nifedipine markedly reduces the induction of c-fos and nur-77 by glucose and GLP-1. Glucose 142-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 9737720-4 1998 The accumulation of c-fos, JunB, and nur-77 mRNA occurs at physiological concentrations of glucose (3 to 11 mM), requires a 1-2 h period, and is mimicked by other nutrient stimuli including mannose, leucine plus glutamine, and pyruvate. Glucose 91-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 9737720-9 1998 The results indicate that GLP-1 and PACAP-38 act as competence factors for the action of glucose on c-fos, JunB, and nur-77. Glucose 89-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 9737720-4 1998 The accumulation of c-fos, JunB, and nur-77 mRNA occurs at physiological concentrations of glucose (3 to 11 mM), requires a 1-2 h period, and is mimicked by other nutrient stimuli including mannose, leucine plus glutamine, and pyruvate. Mannose 190-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 9737720-4 1998 The accumulation of c-fos, JunB, and nur-77 mRNA occurs at physiological concentrations of glucose (3 to 11 mM), requires a 1-2 h period, and is mimicked by other nutrient stimuli including mannose, leucine plus glutamine, and pyruvate. Leucine 199-206 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 9737720-4 1998 The accumulation of c-fos, JunB, and nur-77 mRNA occurs at physiological concentrations of glucose (3 to 11 mM), requires a 1-2 h period, and is mimicked by other nutrient stimuli including mannose, leucine plus glutamine, and pyruvate. Glutamine 212-221 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 9737720-4 1998 The accumulation of c-fos, JunB, and nur-77 mRNA occurs at physiological concentrations of glucose (3 to 11 mM), requires a 1-2 h period, and is mimicked by other nutrient stimuli including mannose, leucine plus glutamine, and pyruvate. Pyruvic Acid 227-235 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 9776515-1 1998 This article is the fifth of a series aimed at mapping brain activities as they result from the development of cyclophosphamide (CP) cystitis in behaving rats using c-fos and Krox-24 expression. Cyclophosphamide 111-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 9743570-5 1998 Audiogenic seizures caused a marked induction of c-fos messenger RNA (mRNA) in septal nucleus, bed nucleus of stria terminalis, amygdaloid nuclei, peripeduncular nucleus, and inferior colliculus, which was almost completely blocked by the pretreatment with MK-801. Dizocilpine Maleate 257-263 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 9731567-0 1998 Chronic alcohol intake reduces retinoic acid concentration and enhances AP-1 (c-Jun and c-Fos) expression in rat liver. Alcohols 8-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 9731567-1 1998 Chronic ethanol intake may interfere with retinoid signal transduction by inhibiting retinoic acid synthesis and by enhancing activator protein-1 (AP-1) (c-Jun and c-Fos) expression, thereby contributing to malignant transformation. Ethanol 8-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-169 9731567-1 1998 Chronic ethanol intake may interfere with retinoid signal transduction by inhibiting retinoic acid synthesis and by enhancing activator protein-1 (AP-1) (c-Jun and c-Fos) expression, thereby contributing to malignant transformation. Retinoids 42-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-169 10027611-12 1998 In the uteri of spayed rats, onapristone was also found to enhance the oestradiol-stimulatory effect on expression of the oestrogen-dependent proto-oncogene, c-fos. onapristone 29-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-163 9731567-8 1998 However, chronic alcohol feeding enhanced AP-1 (c-Jun and c-Fos) expression by 7- to 8-fold, as compared with the control group. Alcohols 17-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 10027611-12 1998 In the uteri of spayed rats, onapristone was also found to enhance the oestradiol-stimulatory effect on expression of the oestrogen-dependent proto-oncogene, c-fos. Estradiol 71-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-163 9710591-1 1998 The proto-oncogenes jun and fos are members of the AP-1 family of transcription factors, which activate transcription of target genes via the tetradecanoyl phorbol acetate response element (TRE). Tetradecanoylphorbol Acetate 142-171 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 9712650-8 1998 NMDA receptors also seem to have a central role in dendrodendritic inhibition in vivo, because intraperitoneal dizocilpine maleate (MK-801) injection in young adult rats resulted in disinhibition of mitral cells as measured by the generation of c-fos mRNA. Dizocilpine Maleate 111-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 245-250 9712650-8 1998 NMDA receptors also seem to have a central role in dendrodendritic inhibition in vivo, because intraperitoneal dizocilpine maleate (MK-801) injection in young adult rats resulted in disinhibition of mitral cells as measured by the generation of c-fos mRNA. Dizocilpine Maleate 132-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 245-250 9804312-3 1998 Manganese-induced apoptosis is accompanied by the induction of DNA fragmentation, expressions of c-Fos and c-Jun, and activation of the c-Jun N-terminal kinase U(JNK) pathway. Manganese 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 9710591-8 1998 Mutations within the C terminus of c-fos at serine residues that are phosphorylation targets for growth factors and MAP kinase completely abrogate transactivation and block potentiation by MAP kinase. Serine 44-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 9710591-9 1998 Using GAL4 fusions, we show that the 90-amino-acid C terminus of c-fos contains autonomous activation domains and that the serine residues are essential for full activity. Serine 123-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 9731622-18 1998 The PC-induced IT is also associated with increased expression of the neuroprotective protein interleukin-1 receptor antagonist and a reduced postischemic expression of the early response genes c-fos and zif268. pc 4-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 194-199 9881874-2 1998 Intradermal injection of carrageenan in the hind-paw produced inflammation in the foot pad, increased the number of cells exhibiting Fos-like immunoreactivity in the dorsal horn of the spinal cord, and decreased the paw-withdrawal latency. Carrageenan 25-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-136 9881874-3 1998 Intradermal injection of capsazepine, a capsaicin-receptor antagonist, significantly reduced the number of cells exhibiting Fos-like immunoreactivity, significantly increased the paw-withdrawal latency, but did not decrease inflammation induced by carrageenan injection. capsazepine 25-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-127 9881874-4 1998 Intradermal injection of capsaicin or formalin also increased Fos-positive neurons. Capsaicin 25-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 9881874-4 1998 Intradermal injection of capsaicin or formalin also increased Fos-positive neurons. Formaldehyde 38-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 9881874-5 1998 Capsaicin- or formalin-induced Fos expression was reduced in both cases by pretreatment of capsazepine, but to a much lesser extent for formalin. Capsaicin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 9881874-5 1998 Capsaicin- or formalin-induced Fos expression was reduced in both cases by pretreatment of capsazepine, but to a much lesser extent for formalin. Formaldehyde 14-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 9881874-5 1998 Capsaicin- or formalin-induced Fos expression was reduced in both cases by pretreatment of capsazepine, but to a much lesser extent for formalin. capsazepine 91-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 9881874-5 1998 Capsaicin- or formalin-induced Fos expression was reduced in both cases by pretreatment of capsazepine, but to a much lesser extent for formalin. Formaldehyde 136-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 9748542-0 1998 Propranolol attenuates haloperidol-induced Fos expression in discrete regions of rat brain: possible brain regions responsible for akathisia. Propranolol 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 9704883-6 1998 We found that selective lesions of serotonergic terminals in the rat forebrain using 5,7-dihydroxytryptamine prevented the full induction of striatal c-Fos by 4 mg/kg amphetamine. 5,7-Dihydroxytryptamine 85-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-155 9704883-6 1998 We found that selective lesions of serotonergic terminals in the rat forebrain using 5,7-dihydroxytryptamine prevented the full induction of striatal c-Fos by 4 mg/kg amphetamine. Amphetamine 167-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-155 9704883-7 1998 Furthermore, amphetamine-induced striatal c-Fos was completely inhibited by administration of the 5-HT3 receptor antagonist, MDL-72222, but not by the 5-HT2A/2C receptor antagonist, ritanserin. Amphetamine 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 9704883-10 1998 These results suggest that 5-HT3 receptor activation may be required for amphetamine-induced expression of ATF-1-regulated target genes in the striatum, which may include c-Fos. Amphetamine 73-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-176 9748483-0 1998 A comparison of delta 9-THC and anandamide induced c-fos expression in the rat forebrain. Dronabinol 16-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 9748483-0 1998 A comparison of delta 9-THC and anandamide induced c-fos expression in the rat forebrain. anandamide 32-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 9748483-4 1998 delta 9-THC and anandamide caused equally high levels of c-fos expression in the paraventricular nucleus of the hypothalamus and the lateral septum. Dronabinol 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 9748483-4 1998 delta 9-THC and anandamide caused equally high levels of c-fos expression in the paraventricular nucleus of the hypothalamus and the lateral septum. anandamide 16-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 9748483-6 1998 Only delta 9-THC caused significant increases in c-fos expression in the nucleus accumbens and caudate-putamen. Dronabinol 5-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 9729312-0 1998 Brain fos-like immunoreactivity in chronic decerebrate and neurologically intact rats given 2,5-anhydro-D-mannitol. 2,5-anhydromannitol 92-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 6-9 9729312-1 1998 Injection of the fructose analogue, 2,5-anhydro-d-mannitol (2,5-AM), increases food intake and Fos-like immunoreactivity (Fos-li) in both brainstem and forebrain structures. Fructose 17-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 9729312-1 1998 Injection of the fructose analogue, 2,5-anhydro-d-mannitol (2,5-AM), increases food intake and Fos-like immunoreactivity (Fos-li) in both brainstem and forebrain structures. Fructose 17-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 9729312-1 1998 Injection of the fructose analogue, 2,5-anhydro-d-mannitol (2,5-AM), increases food intake and Fos-like immunoreactivity (Fos-li) in both brainstem and forebrain structures. 2,5-anhydromannitol 36-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 9729312-1 1998 Injection of the fructose analogue, 2,5-anhydro-d-mannitol (2,5-AM), increases food intake and Fos-like immunoreactivity (Fos-li) in both brainstem and forebrain structures. 2,5-anhydromannitol 36-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 9729312-1 1998 Injection of the fructose analogue, 2,5-anhydro-d-mannitol (2,5-AM), increases food intake and Fos-like immunoreactivity (Fos-li) in both brainstem and forebrain structures. 2,5-anhydromannitol 60-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 9729312-1 1998 Injection of the fructose analogue, 2,5-anhydro-d-mannitol (2,5-AM), increases food intake and Fos-like immunoreactivity (Fos-li) in both brainstem and forebrain structures. 2,5-anhydromannitol 60-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 9729312-6 1998 Both intact and CD rats showed increased Fos-li in the nucleus of the solitary tract (NTS) after injection of 2,5-AM as compared with saline. Cadmium 16-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 9729312-6 1998 Both intact and CD rats showed increased Fos-li in the nucleus of the solitary tract (NTS) after injection of 2,5-AM as compared with saline. 2,5-anhydromannitol 110-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 9729312-7 1998 2, 5-AM treatment increased Fos-li in the external lateral division of parabrachial nucleus (PBNel) in intact but not in CD rats, suggesting that descending projections from the forebrain may play a role in the activation of PBNel neurons after 2,5-AM injection. 2, 5-am 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 9729312-8 1998 Decerebration eliminated significant 2,5-AM-induced Fos-li responses in forebrain structures, including the paraventricular nucleus, supraoptic nucleus, bed nucleus of the stria terminalis and central nucleus of the amygdala. 2,5-anhydromannitol 37-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 9729361-0 1998 Central infusion of glucagon-like peptide-1-(7-36) amide (GLP-1) receptor antagonist attenuates lithium chloride-induced c-Fos induction in rat brainstem. Amides 51-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-126 9729361-0 1998 Central infusion of glucagon-like peptide-1-(7-36) amide (GLP-1) receptor antagonist attenuates lithium chloride-induced c-Fos induction in rat brainstem. Lithium Chloride 96-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-126 9729361-2 1998 injection of lithium chloride (LiCl) produce similar patterns of c-Fos induction in the rat brain. Lithium Chloride 13-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 9729361-2 1998 injection of lithium chloride (LiCl) produce similar patterns of c-Fos induction in the rat brain. Lithium Chloride 31-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 9729361-5 1998 We therefore determined if third-ventricular (i3vt) infusion of a GLP-1 receptor antagonist would block LiCl-induced c-Fos expression in the brainstem. Lithium Chloride 104-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 9729361-9 1998 These results suggest that LiCl-induced c-Fos expression in the rat brainstem is mediated, at least in part, by GLP-1 receptor signaling. Lithium Chloride 27-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 9688679-4 1998 The effects of these inhibitors on brain Fos-li in several specific brain nuclei paralleled those on feeding behavior; that is, the number of cells showing Fos-li increased only under dietary conditions in which 2,5-AM or MP stimulated eating. 2,5-anhydromannitol 212-218 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 9688679-4 1998 The effects of these inhibitors on brain Fos-li in several specific brain nuclei paralleled those on feeding behavior; that is, the number of cells showing Fos-li increased only under dietary conditions in which 2,5-AM or MP stimulated eating. 2,5-anhydromannitol 212-218 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-159 9748542-0 1998 Propranolol attenuates haloperidol-induced Fos expression in discrete regions of rat brain: possible brain regions responsible for akathisia. Haloperidol 23-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 9748542-5 1998 To identify the neural substrates of akathisia, we investigated the effects of propranolol on haloperidol-induced Fos expression in rat brain. Propranolol 79-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 9748542-5 1998 To identify the neural substrates of akathisia, we investigated the effects of propranolol on haloperidol-induced Fos expression in rat brain. Haloperidol 94-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 9748542-6 1998 Haloperidol (1 mg/kg) induced Fos-positive nuclei in several regions of the brain, including the cingulate cortex area 3, piriform cortex nucleus accumbens, caudate-putamen, ventral lateral septum and parietal cortex. Haloperidol 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 9748542-7 1998 Pretreatment with propranolol (5 mg/kg) reduced the number of Fos-positive nuclei in the cingulate cortex area 3, the piriform cortex and area 1 of the parietal cortex. Propranolol 18-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 9681493-6 1998 A MAPK kinase inhibitor (PD98059) also reduced the AM-induced MAPK activation, c-fos messenger RNA expression, and cell proliferation. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 25-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 11324543-0 1998 [Neurokinin-1 receptor mediated formalin-induced c-fos expression in the rat spinal cord]. Formaldehyde 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 20654422-1 1998 In the present study, the time- and concentration-dependent effects of the carcinogen benzo[a]pyrene (BaP) on c-fos, c-jun, c-myc and c-Ha-ras expression were evaluated in primary cultured rat hepatocytes. Benzo(a)pyrene 86-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 20654422-6 1998 Challenge with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) (1 nm), an aryl hydrocarbon receptor (AhR) agonist, elicited responses comparable to BaP for c-jun and c-Ha-ras, but not for c-fos or c-myc. Polychlorinated Dibenzodioxins 52-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 183-188 9764482-4 1998 An L-type calcium channel blocker, nifedipine, also inhibited the electrically induced calcium influx and c-fos expression. Nifedipine 35-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 9764482-4 1998 An L-type calcium channel blocker, nifedipine, also inhibited the electrically induced calcium influx and c-fos expression. Calcium 10-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 9671276-7 1998 The number of c-Fos-LI neurons increased dose dependently (r = 0.973, n = 24) for carrageenan, from a number close to zero for the saline injection. Sodium Chloride 131-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 9671276-8 1998 At the PB level, c-Fos was predominantly expressed contralateral to intraplantar carrageenan. pladienolide B 7-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 9671276-8 1998 At the PB level, c-Fos was predominantly expressed contralateral to intraplantar carrageenan. Carrageenan 81-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 9671276-10 1998 The number of c-Fos-LI neurons in the PB area was correlated with that in the superficial laminae (r = 0.935, n = 24) and with the paw edema (r = 0.931, n = 24). pladienolide B 38-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 9721035-0 1998 Olanzapine-induced Fos expression in the rat forebrain; cross-tolerance with haloperidol and clozapine. Olanzapine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 9721035-0 1998 Olanzapine-induced Fos expression in the rat forebrain; cross-tolerance with haloperidol and clozapine. Haloperidol 77-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 9721035-3 1998 The acutely-induced Fos responses of olanzapine were significantly reduced in all brain areas investigated after a 3-week treatment period, indicating the development of tolerance. Olanzapine 37-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 9721035-4 1998 Through evaluation of cross-tolerance we investigated whether the effects of olanzapine, haloperidol and clozapine on Fos expression and on plasma corticosterone are mediated by the same or by different mechanisms. Clozapine 105-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 9651224-2 1998 The purpose of the present study was to investigate the possible supraspinal contribution of ABT-594 by assessing its ability to induce expression of the immediate early gene c-fos, a biochemical marker of neuronal activation, in the NRM of rats. 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid 93-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 175-180 9651224-5 1998 produced a dose-dependent induction of Fos protein that was blocked by the central nicotinic acetylcholine receptor (nAChR) antagonist mecamylamine (5 micromol/kg, i.p.) Mecamylamine 135-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 9666120-0 1998 Expression of c-fos and hsp70 mRNA in neonatal rat cerebrocortical slices during NMDA-induced necrosis and apoptosis. N-Methylaspartate 81-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 9666163-0 1998 Acute ethanol induces c-fos immunoreactivity in GABAergic neurons of the central nucleus of the amygdala. Ethanol 6-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 9666163-2 1998 Previous studies have shown that acute ethanol administration induces the expression of c-fos in the CNA of rat brains. Ethanol 39-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 9720795-17 1998 Reverse transcription-polymerase chain reaction analysis of c-fos, c-jun and c-myc mRNA levels demonstrated that c-jun mRNA level after serum-stimulation was significantly reduced by 10(-5) M EGC. gallocatechol 192-195 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 9720795-18 1998 However, the reduction of c-fos and c-myc mRNA levels by 10(-5) M EGC did not achieve significance. gallocatechol 66-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 9754718-5 1998 We found that trifluoperazine inhibited the expression of the H+-induced c-fos/c-jun mRNA by 30-35%. Trifluoperazine 14-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 9655894-2 1998 Here, we report that alpha-1 B ARs inhibit the activities of alpha-1A ARs in neonatal rat myocardium so that the inactivation of alpha-1 B ARs by chloroethylclonidine (CEC) potentiated the effects of nonselective alpha-1 AR agonist phenylephrine (PE) on myocardial protein synthesis and early gene (c-fos and c-jun) expression. chlorethylclonidine 146-166 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 299-304 9655894-2 1998 Here, we report that alpha-1 B ARs inhibit the activities of alpha-1A ARs in neonatal rat myocardium so that the inactivation of alpha-1 B ARs by chloroethylclonidine (CEC) potentiated the effects of nonselective alpha-1 AR agonist phenylephrine (PE) on myocardial protein synthesis and early gene (c-fos and c-jun) expression. chlorethylclonidine 168-171 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 299-304 9655895-5 1998 Accompanying behavioral sensitization were two postsynaptic neuroadaptive responses: reduction in the ability of Amp to induce c-fos gene expression in the infralimbic/ventral prelimbic cortex and NT/N mRNA in the accumbal shell. Amphetamine 113-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 9655895-6 1998 However, concurrent blockade of D4 receptors during Amp pretreatment prevented the refractoriness in c-fos and NT/N responsiveness to acute Amp. Amphetamine 52-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 9608582-4 1998 We examined the effect of acute administration of desmethylclozapine to rats on forebrain Fos protein expression. norclozapine 50-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 9622249-2 1998 A rapid and transient increase in c-fos, junB, c-jun and Tis11 messenger RNA was observed in cultured astrocytes after treatment with adenosine-5"-O-(2-thiodiphosphate). adenosine-5"-o-( 134-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 9622249-2 1998 A rapid and transient increase in c-fos, junB, c-jun and Tis11 messenger RNA was observed in cultured astrocytes after treatment with adenosine-5"-O-(2-thiodiphosphate). 2-thiodiphosphate 150-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 9622249-5 1998 The combined stimulation of astrocytes with calcitonin gene-related peptide and adenosine-5"-O-(2-thiodiphosphate) resulted in the potentiated expression of c-fos messenger RNA. adenosine 5'-O-(2-thiodiphosphate) 80-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-162 9718284-0 1998 Induction of Fos protein by antipsychotic drugs in rat brain following kainic acid-induced limbic-cortical neuronal loss. Kainic Acid 71-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 9718284-3 1998 infusion of kainic acid (KA) produces loss of limbic-cortical neurons that project to these brain areas, we postulated that the c-fos responses to antipsychotics in these brain areas would be altered following i.c.v. Kainic Acid 12-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-133 9718284-8 1998 In both KA-lesioned and control animals, haloperidol produced greater increases in Fos protein immunoreactivity in the striatum than in limbic-cortical areas, while clozapine produced greater increases in Fos protein immunoreactivity in limbic-cortical areas than in the striatum. Haloperidol 41-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 9718284-8 1998 In both KA-lesioned and control animals, haloperidol produced greater increases in Fos protein immunoreactivity in the striatum than in limbic-cortical areas, while clozapine produced greater increases in Fos protein immunoreactivity in limbic-cortical areas than in the striatum. Clozapine 165-174 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 205-208 9718284-9 1998 In both KA-lesioned and control animals, haloperidol and clozapine administration also produced similar dose-dependent increases in Fos protein immunoreactivity in the striatum and nucleus accumbens. Haloperidol 41-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-135 9718284-9 1998 In both KA-lesioned and control animals, haloperidol and clozapine administration also produced similar dose-dependent increases in Fos protein immunoreactivity in the striatum and nucleus accumbens. Clozapine 57-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-135 9718284-10 1998 However, the ability of clozapine to increase Fos protein immunoreactivity in the infralimbic prefrontal cortex was significantly enhanced in KA-lesioned rats compared to controls. Clozapine 24-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 9718284-11 1998 Since limbic-cortical pathology has been implicated in the negative symptoms of schizophrenia, the enhanced effect of clozapine on limbic-cortical expression of c-fos in KA-lesioned rats may be relevant to understanding clozapine"s unusual therapeutic actions in patients with schizophrenia. Clozapine 118-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 9718284-11 1998 Since limbic-cortical pathology has been implicated in the negative symptoms of schizophrenia, the enhanced effect of clozapine on limbic-cortical expression of c-fos in KA-lesioned rats may be relevant to understanding clozapine"s unusual therapeutic actions in patients with schizophrenia. Clozapine 220-229 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 10375794-0 1998 [Effect of corticotropin on formalin-evoked c-fos expression of spinal cord and receptors analysis in rat]. Formaldehyde 28-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 10375794-1 1998 AIM: To study the effect of corticotropin (Cor) on c-fos expression induced by formalin in spinal cord of rat and the role of receptors. Formaldehyde 79-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 10375794-3 1998 RESULTS: Cor inhibited the formalin-evoked c-fos expression in rat spinal cord in a dose-dependent manner. Formaldehyde 27-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 10375794-5 1998 Cor 10 U.kg-1, but 25 U.kg-1 reduced the evoked c-fos expression, that was blocked by phentolamine, naloxone, or verapamil, but not much changed by adrenalectomy. Phentolamine 86-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 10375794-5 1998 Cor 10 U.kg-1, but 25 U.kg-1 reduced the evoked c-fos expression, that was blocked by phentolamine, naloxone, or verapamil, but not much changed by adrenalectomy. Naloxone 100-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 10375794-5 1998 Cor 10 U.kg-1, but 25 U.kg-1 reduced the evoked c-fos expression, that was blocked by phentolamine, naloxone, or verapamil, but not much changed by adrenalectomy. Verapamil 113-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 10375794-6 1998 CONCLUSION: The formalin-evoked c-fos expression of rat spinal cord was suppressed by Cor through the alpha-adrenergic receptors, opiate receptors and calcium, but no relation to adrenal glands. Formaldehyde 16-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 9647745-0 1998 Evidence for role of phospholipase A2 in phosphatidic acid-induced signaling to c-fos serum response element activation. Phosphatidic Acids 41-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 9647745-1 1998 The activity of exogenous phosphatidic acid (PA) to transactivate c-fos serum response element (SRE) was investigated by transient transfection analysis. Phosphatidic Acids 26-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 9647745-1 1998 The activity of exogenous phosphatidic acid (PA) to transactivate c-fos serum response element (SRE) was investigated by transient transfection analysis. Phosphatidic Acids 45-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 9647745-2 1998 Incubation of Rat-2 fibroblast cells with exogenous PA caused a stimulation of c-fos SRE-linked luciferase activity in a dose- and time-dependent manner. Phosphatidic Acids 52-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 9716316-0 1998 Patterns of c-fos expression induced by fluvoxamine are different after acute vs. chronic oral administration. Fluvoxamine 40-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 9716316-2 1998 We used the expression of c-fos, after both acute and chronic oral administration of fluvoxamine in the rat, to study its immediate and long-term effects, in relation to the distribution of Galanin (GAL) and Vasoactive Intestinal Polypeptide (VIP). Fluvoxamine 85-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 9716316-5 1998 It is concluded that activation of 5-HT3-receptors in the caudal brainstem or gastro-intestinal afferents of the vagal nerve may play a role in the observed pattern of Fos-IR after fluvoxamine administration. Fluvoxamine 181-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-171 9826788-2 1998 In the present study, treatment of cultured rat embryonic basal forebrain neurons with anti-c-fos, prior to administering NGF, blocked NGF-mediated increases in ChAT activity by 67%; basal ChAT activity was not affected by the antisense oligonucleotide treatment. Oligonucleotides 237-252 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 9681947-1 1998 The number and distribution of Fos-like-immunoreactive neurons in different supraspinal brain areas induced by formalin injection into one hindpaw was estimated in rats with transected dorsal half of the spinal cord at the thoracic level in an attempt to avoid most of the descending modulatory actions. Formaldehyde 111-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 9661252-0 1998 Inducibility of c-Fos protein in visuo-motor system and limbic structures after acute and repeated administration of nicotine in the rat. Nicotine 117-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 9661252-1 1998 To identify neuroanatomical substrates affected by nicotine, we have studied its effects after acute and repeated administration through the c-Fos protein inducibility in various brain structures. Nicotine 51-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 9661252-2 1998 Ninety minutes after acute nicotine (0.35 mg/kg, s.c.) the number of c-Fos-like immunoreactive nuclei was consistently increased in visuo-motor structures such as the superior colliculus, the medial terminal nucleus of accessory optic tract, and the nucleus of the optic tract. Nicotine 27-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 9661252-5 1998 In chronically treated rats (0.35 mg/kg s.c., 3 x day for 14 days), the last nicotine injection given on the 15th day was still able to induce 90 minutes later c-Fos protein in visuo-motor, retino-limbic, subcortical, and cortical limbic structures. Nicotine 77-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 9661252-7 1998 c-Fos induction after nicotine differs from that reported after other addictive drugs in terms of pattern and chronic inducibility, indicating that different mechanisms are involved for maintaining this transcription factor. Nicotine 22-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 11324543-1 1998 In the present work, the effects of tachykinin receptor antagonists on formalin-induced c-fos expression in the rat spinal cord were studied by a combination of immunocytochemical and neuropharmacological methods. Formaldehyde 71-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 11324543-2 1998 Formalin injected into a hindpaw of rat only resulted in c-fos expression in the ipsilateral dorsal horn neurons of the spinal cord. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 11324543-6 1998 Our results indicated that NK-1 receptor mediated formalin-induced c-fos expression in the spinal cord, suggesting NK-1 receptor might play an important role in the spinal transmission of nociceptive messages. Formaldehyde 50-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 9668442-0 1998 Cocaine-inhibited neuronal differentiation in NGF-induced PC12 cells and altered c-fos expression are reversed by C-fos antisense oligonucleotide. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 9668442-0 1998 Cocaine-inhibited neuronal differentiation in NGF-induced PC12 cells and altered c-fos expression are reversed by C-fos antisense oligonucleotide. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 9668442-0 1998 Cocaine-inhibited neuronal differentiation in NGF-induced PC12 cells and altered c-fos expression are reversed by C-fos antisense oligonucleotide. Oligonucleotides 130-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 9668442-0 1998 Cocaine-inhibited neuronal differentiation in NGF-induced PC12 cells and altered c-fos expression are reversed by C-fos antisense oligonucleotide. Oligonucleotides 130-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 9696058-0 1998 Globus pallidus lesions inhibit the induction of c-Fos by haloperidol in the basal ganglia output nuclei in rats. Haloperidol 58-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 9630573-0 1998 Expression of fos protein in rat brain following administration of a nicotinic acetylcholine receptor agonist epibatidine. epibatidine 110-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 9630573-2 1998 The present study was undertaken to examine the expression of Fos protein in several rat brain regions following an acute administration of epibatidine. epibatidine 140-151 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 9630573-3 1998 Furthermore, we also studied the role of the dopamine D1 and D2 receptors and the N-methyl-d-aspartate (NMDA) receptor, and nicotinic ACh receptor in the expression of Fos protein by epibatidine. epibatidine 183-194 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-171 9630573-4 1998 A single administration of epibatidine (5, 10, 50 microgram/kg) caused a marked induction of Fos-immunoreactivity in the prefrontal cortex, medial striatum, nucleus accumbens, amygdala and superior colliculus of rat brain. epibatidine 27-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 9630573-7 1998 Pretreatment with sulpiride, a dopamine D2 receptor antagonist, blocked the induction of Fos protein in the prefrontal cortex and the core region of accumbens nucleus, but not in the medial striatum and the shell division of nucleus accumbens of rat brain. Sulpiride 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 9630573-8 1998 These results suggest that epibatidine induced the expression of Fos protein in several regions of rat brain, and that dopamine D1 receptor, NMDA receptor, and nicotinic ACh receptor may play a role in the expression of Fos protein by epibatidine in rat brain. epibatidine 27-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 9630573-8 1998 These results suggest that epibatidine induced the expression of Fos protein in several regions of rat brain, and that dopamine D1 receptor, NMDA receptor, and nicotinic ACh receptor may play a role in the expression of Fos protein by epibatidine in rat brain. epibatidine 27-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 220-223 9630573-8 1998 These results suggest that epibatidine induced the expression of Fos protein in several regions of rat brain, and that dopamine D1 receptor, NMDA receptor, and nicotinic ACh receptor may play a role in the expression of Fos protein by epibatidine in rat brain. epibatidine 235-246 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 220-223 9630573-9 1998 Furthermore, dopamine D2 receptor may, in part, play a role in epibatidine induced expression of Fos protein in the prefrontal cortex and the core region of nucleus accumbens, but not in the medial striatum and the shell division of nucleus accumbens of rat brain. epibatidine 63-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 9662427-4 1998 Incubation with RA did not alter NGFI-A mRNA levels, but significantly reduced the NGFI-B and c-fos response to NGF and serum. Tretinoin 16-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 9662427-6 1998 Sequences contained within the 5" flanking region of the c-fos gene confer responsiveness to NGF and mediate the inhibitory effect of RA. Tretinoin 134-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 9666136-0 1998 Arachidonylethanolamide (AEA) activation of FOS proto-oncogene protein immunoreactivity in the rat brain. anandamide 0-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 9666136-0 1998 Arachidonylethanolamide (AEA) activation of FOS proto-oncogene protein immunoreactivity in the rat brain. anandamide 25-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 9666136-4 1998 In this study, FOS immunoreactivity (FOSir) was used to map rat brain nuclei that are responsive to a single intracerebroventricular injection of AEA. anandamide 146-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-18 9633917-8 1998 Pretreatment of myocytes with staurosporine also reduced the myotrophin-induced mRNA levels of c-fos and beta-myosin heavy chain. Staurosporine 30-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 9696058-1 1998 This study examined the induction of c-Fos expression in the substantia nigra pars reticulata (SNr) and entopeduncular nucleus (EP) in the rats with a globus pallidus (GP) lesion, following the administration of haloperidol. Haloperidol 212-223 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 9622600-6 1998 In adult rats, systemic injection of kainic acid induced c-fos expression in granule cells and stellate/basket interneurons within 1 h of treatment. Kainic Acid 37-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 9696058-3 1998 Haloperidol induced a high level of the expression of c-Fos in neurons of the SNr and EP, and the GP lesion significantly decreased the expression of c-Fos in the ipsilateral SNr and EP. Haloperidol 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 9622600-9 1998 A combination of lesioning and systemic kainic acid produced a strong, c-fos expression throughout the ipsilateral granular layer as well as in Purkinje cell nuclei. Kainic Acid 40-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 9622600-11 1998 We conclude that c-fos gene expression occurs transiently in granule cells during postnatal development and can be rapidly re-induced in the adult following systemic injection of glutamate agonists. Glutamic Acid 179-188 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 9696058-4 1998 Since it has been suggested that c-Fos expression in the SNr/EP is caused by increased excitatory inputs from the subthalamic nucleus (STN), the present results provide functional evidence indicating that neuronal activities of the basal ganglia output nuclei are not increased by GP ablation, unlike D2 receptor blockade, supporting the recently proposed hypothesis that overactivity of the STN resulting from dopamine depletion is not solely a result of disinhibition from inhibitory GP efferents. Dopamine 411-419 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 29087601-0 1998 Cocaine-Inhibited Neuronal Differentiation in NGF-Induced PC12 Cells and Altered C-fos Expression Are Reversed by C-fos Antisense Oligonucleotidea. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 29087601-0 1998 Cocaine-Inhibited Neuronal Differentiation in NGF-Induced PC12 Cells and Altered C-fos Expression Are Reversed by C-fos Antisense Oligonucleotidea. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 29087601-0 1998 Cocaine-Inhibited Neuronal Differentiation in NGF-Induced PC12 Cells and Altered C-fos Expression Are Reversed by C-fos Antisense Oligonucleotidea. oligonucleotidea 130-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 29087601-0 1998 Cocaine-Inhibited Neuronal Differentiation in NGF-Induced PC12 Cells and Altered C-fos Expression Are Reversed by C-fos Antisense Oligonucleotidea. oligonucleotidea 130-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 9630593-0 1998 c-fos gene expression is induced in a subpopulation of striatal neurons following a single administration of a dopamine D1-receptor agonist in adult rats lesioned with 6-OHDA as neonates. Oxidopamine 168-174 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9841547-5 1998 Linoleic acid suppressed intake and stimulated Fos expression similarly to glucose infusions of three times the caloric value. Linoleic Acid 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 9841553-4 1998 Furo/Cap treatment significantly increased Fos-ir density above baseline levels both in structures of the lamina terminalis and hypothalamus known to mediate the actions of ANG II and in hindbrain regions associated with blood volume and pressure regulation. furo 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 9841553-4 1998 Furo/Cap treatment significantly increased Fos-ir density above baseline levels both in structures of the lamina terminalis and hypothalamus known to mediate the actions of ANG II and in hindbrain regions associated with blood volume and pressure regulation. cap 5-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 9841553-5 1998 Furo/Cap treatment also typically increased Fos-ir density in these structures above levels observed after administration of furosemide or captopril separately. furo 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 9841553-5 1998 Furo/Cap treatment also typically increased Fos-ir density in these structures above levels observed after administration of furosemide or captopril separately. cap 5-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 9841553-6 1998 Fos-ir was reduced to a greater extent in forebrain than in hindbrain areas by a dose of captopril (100 mg/kg sc) known to block the actions of ACE in the brain. Captopril 89-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9590563-0 1998 Differential expression of c-fos mRNA and Fos protein in the rat brain after restraint stress or pentylenetetrazol-induced seizures. Pentylenetetrazole 97-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 9590563-0 1998 Differential expression of c-fos mRNA and Fos protein in the rat brain after restraint stress or pentylenetetrazol-induced seizures. Pentylenetetrazole 97-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 9590563-5 1998 Forced restraint stress and PTZ-induced seizures generated c-fos mRNA expression of distinct intensities, but in similar brain regions, including the hippocampus, the amygdala, the piriform cortex, the paraventricular hypothalamic nucleus, the habenula, and parts of the cerebral cortex. Pentylenetetrazole 28-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 9590563-9 1998 Nevertheless, both areas presented Fos-like expression after PTZ-induced seizures. Pentylenetetrazole 61-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 9630593-1 1998 The effects of the dopamine D1 receptor agonist, SKF-38393, on the levels of mRNAs encoding for the proto-oncogene c-fos and the GABA-synthesizing enzyme glutamate decarboxylase (GAD65) were measured by in situ hybridization histochemistry in the striatum of adult rats depleted of dopamine as neonates. Dopamine 19-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-120 9630593-2 1998 c-fos mRNA levels exhibited a prominent increase following the acute systemic administration of SKF-38393 in dopamine-depleted but not in normal rats. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 96-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9630593-2 1998 c-fos mRNA levels exhibited a prominent increase following the acute systemic administration of SKF-38393 in dopamine-depleted but not in normal rats. Dopamine 109-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9630593-3 1998 Double-labeling in situ hybridization histochemistry using a radioactive c-fos probe and a digoxigenin-labeled preproenkephalin (PPE) cRNA probe indicated that c-fos mRNA levels were increased by SKF-38393 exclusively in a subpopulation of PPE-unlabeled neurons. Digoxigenin 91-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 9607780-0 1998 The xenoestrogen bisphenol A induces growth, differentiation, and c-fos gene expression in the female reproductive tract. bisphenol A 17-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 9698833-5 1998 In the caudal sub-nucleus of the spinal trigeminal nuclear complex, the number of Fos-positive neurons was much higher in rats with inflammation of the superior cervical ganglion than in control rats, either sham-operated or with saline applied to the ganglion. Sodium Chloride 230-236 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 9655238-4 1998 Expression of c-fos in brain regions sensitive to kainic acid was quicker but lasted a noticeably shorter time in microencephalic rats as compared to normal animals. Kainic Acid 50-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 9661807-10 1998 Lovastatin as well as calphostin C partially but significantly inhibited the stretch-induced increases in MAP kinase activity, c-fos mRNA expression, and protein synthesis. Lovastatin 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 9626636-6 1998 We also found that while inhibition of either PG or NO production at the time of loading caused a partial suppression of c-fos mRNA expression in the loaded vertebrae, administration of indomethacin and NG-monomethyl-L-arginine together markedly suppressed c-fos expression. Prostaglandins 46-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-126 9626636-6 1998 We also found that while inhibition of either PG or NO production at the time of loading caused a partial suppression of c-fos mRNA expression in the loaded vertebrae, administration of indomethacin and NG-monomethyl-L-arginine together markedly suppressed c-fos expression. Prostaglandins 46-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 257-262 9626636-6 1998 We also found that while inhibition of either PG or NO production at the time of loading caused a partial suppression of c-fos mRNA expression in the loaded vertebrae, administration of indomethacin and NG-monomethyl-L-arginine together markedly suppressed c-fos expression. omega-N-Methylarginine 203-227 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-126 9626636-6 1998 We also found that while inhibition of either PG or NO production at the time of loading caused a partial suppression of c-fos mRNA expression in the loaded vertebrae, administration of indomethacin and NG-monomethyl-L-arginine together markedly suppressed c-fos expression. omega-N-Methylarginine 203-227 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 257-262 9592116-1 1998 We previously reported that withdrawal from morphine induces the expression of Fos, a marker of neuronal activity, in spinal cord neurons, particularly in laminae I and II of the superficial dorsal horn, and that the magnitude of Fos expression is increased in rats with a midthoracic spinal transection. Morphine 44-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 9592116-1 1998 We previously reported that withdrawal from morphine induces the expression of Fos, a marker of neuronal activity, in spinal cord neurons, particularly in laminae I and II of the superficial dorsal horn, and that the magnitude of Fos expression is increased in rats with a midthoracic spinal transection. Morphine 44-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 230-233 9579785-6 1998 The ability of PACAP to stimulate c-fos gene expression was mimicked by dibutyryladenosine 3",5"-cyclic-monophosphate but not phorbol 12-myristate 13-acetate. Bucladesine 72-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 9681590-0 1998 Widespread expression of Fos protein induced by acute haloperidol administration in the rat brain. Haloperidol 54-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 9681590-1 1998 The effect of acute haloperidol administration on Fos protein expression was examined immunohistochemically in discrete regions of the rat brain. Haloperidol 20-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 9681590-8 1998 The distribution of changes in Fos immunoreactivity at the high dose of haloperidol (1.0 mg/kg) were comparable to their distribution at the moderate dose. Haloperidol 72-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 9681590-9 1998 These findings indicate that the effect of acute haloperidol on Fos expression is widely distributed in the rat brain beyond the previously known dopamine-rich areas at the dose which produces plasma levels equivalent to those within the therapeutic range used clinically in humans. Haloperidol 49-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 9681590-9 1998 These findings indicate that the effect of acute haloperidol on Fos expression is widely distributed in the rat brain beyond the previously known dopamine-rich areas at the dose which produces plasma levels equivalent to those within the therapeutic range used clinically in humans. Dopamine 146-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 9683006-10 1998 Test day treatment with U-50,488 stimulated Fos IR in various brain regions of the preweanling rat, including the medial striatum, nucleus accumbens, lateral habenula, and septal area. u-50 24-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 9683006-11 1998 Chronic treatment with U-50,488 depressed Fos expression in a number of brain regions (relative to acutely treated rats); however, these changes in Fos IR did not necessarily coincide with the occurrence of behavioral sensitization. u-50 23-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 9683006-11 1998 Chronic treatment with U-50,488 depressed Fos expression in a number of brain regions (relative to acutely treated rats); however, these changes in Fos IR did not necessarily coincide with the occurrence of behavioral sensitization. u-50 23-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-151 9683006-13 1998 Therefore, while acute treatment with U-50,488 both increased locomotor activity and stimulated Fos IR in preweanling rats, chronic U-50,488 treatment produced behavioral changes that did not correspond with Fos expression. u-50 38-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-99 9668660-0 1998 The serotonergic system modulates the cocaine-induced expression of the immediate early genes egr-1 and c-fos in rat brain. Cocaine 38-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 9668660-6 1998 Cocaine-induced egr-1 and c-fos expression was substantially reduced in the brain areas from rats in which the serotonergic projections were lesioned by injection of the neurotoxin 5,7-dihydroxytryptamine and in rats that have been injected with tropisetron, an antagonist of the 5-hydroxytryptamine (5-HT3) receptor. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 9668660-6 1998 Cocaine-induced egr-1 and c-fos expression was substantially reduced in the brain areas from rats in which the serotonergic projections were lesioned by injection of the neurotoxin 5,7-dihydroxytryptamine and in rats that have been injected with tropisetron, an antagonist of the 5-hydroxytryptamine (5-HT3) receptor. 5,7-Dihydroxytryptamine 181-204 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 12016907-0 1998 [Effect of dl-3-N-butylphthalide on the expression of hsp70 mRNA and c-fos in transient cerebral ischemic and reperfused rat brain]. 3-n-butylphthalide 11-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 9630536-0 1998 Stimulant-mediated c-fos induction in striatum as a function of age, sex, and prenatal cocaine exposure. Cocaine 87-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 9630536-1 1998 Induction of the immediate-early gene c-fos by the stimulants cocaine and amphetamine (AMPH) was analyzed by Fos immunocytochemistry at different ages in the brains of prenatally cocaine-treated and control rats. Cocaine 62-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 9630536-1 1998 Induction of the immediate-early gene c-fos by the stimulants cocaine and amphetamine (AMPH) was analyzed by Fos immunocytochemistry at different ages in the brains of prenatally cocaine-treated and control rats. Amphetamine 74-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 9630536-1 1998 Induction of the immediate-early gene c-fos by the stimulants cocaine and amphetamine (AMPH) was analyzed by Fos immunocytochemistry at different ages in the brains of prenatally cocaine-treated and control rats. Amphetamine 87-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 9630536-1 1998 Induction of the immediate-early gene c-fos by the stimulants cocaine and amphetamine (AMPH) was analyzed by Fos immunocytochemistry at different ages in the brains of prenatally cocaine-treated and control rats. Cocaine 179-186 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 9630536-2 1998 Cocaine and AMPH induced c-fos in patches of striatal neurons during the first postnatal week, and thereafter produced a progressively more homogeneous pattern that was more dense medially. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 9630536-2 1998 Cocaine and AMPH induced c-fos in patches of striatal neurons during the first postnatal week, and thereafter produced a progressively more homogeneous pattern that was more dense medially. Amphetamine 12-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 9630536-3 1998 Quantification of Fos-immunoreactive cells in older rats revealed differences related to sex and prenatal cocaine treatment. Cocaine 106-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 9630536-4 1998 Both cocaine and AMPH produced dose-dependent increases in the number of Fos-immunoreactive cells in striatum. Cocaine 5-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-76 9630536-4 1998 Both cocaine and AMPH produced dose-dependent increases in the number of Fos-immunoreactive cells in striatum. Amphetamine 17-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-76 9630536-5 1998 Prenatal cocaine exposure resulted in increased Fos in males in response to AMPH (2 mg/kg) at P18 and cocaine (10 mg/kg) at 1-2 months. Cocaine 9-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 9630536-5 1998 Prenatal cocaine exposure resulted in increased Fos in males in response to AMPH (2 mg/kg) at P18 and cocaine (10 mg/kg) at 1-2 months. Amphetamine 76-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 9630536-5 1998 Prenatal cocaine exposure resulted in increased Fos in males in response to AMPH (2 mg/kg) at P18 and cocaine (10 mg/kg) at 1-2 months. Cocaine 102-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 9630536-8 1998 The dopamine D1 antagonist SCH23390 blocked cocaine-mediated c-fos induction in all groups. SCH 23390 27-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 9630536-8 1998 The dopamine D1 antagonist SCH23390 blocked cocaine-mediated c-fos induction in all groups. Cocaine 44-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 9630536-9 1998 The NMDA antagonist MK-801 blocked cocaine-mediated c-fos induction in the medial striatum. N-Methylaspartate 4-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 9630536-9 1998 The NMDA antagonist MK-801 blocked cocaine-mediated c-fos induction in the medial striatum. Dizocilpine Maleate 20-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 9630536-9 1998 The NMDA antagonist MK-801 blocked cocaine-mediated c-fos induction in the medial striatum. Cocaine 35-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 9630536-10 1998 In females only, MK-801 pretreatment resulted in a dramatic increase in the number of Fos-immunoreactive cells in lateral striatum. Dizocilpine Maleate 17-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 9630536-11 1998 These findings indicate differences in the neural basis of c-fos induction in males and females, and changes in stimulant-mediated c-fos induction resulting from prenatal cocaine exposure. Cocaine 171-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-136 9590559-8 1998 After metrazol-induced seizures, Fos-positive cells were present in CA1 heterotopias, the only hippocampal region to be activated with the neocortex. Pentylenetetrazole 6-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 9630518-8 1998 In conclusion, the present study shows that brain regions with the highest expression of Fos and the largest metabolic activation were also highly stained with acid fuchsin and most heavily damaged. acid fuchsin 160-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 9570817-0 1998 Nerve gas-induced seizures: role of acetylcholine in the rapid induction of Fos and glial fibrillary acidic protein in piriform cortex. Acetylcholine 36-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 9570817-1 1998 Soman (pinacolymethylphosphonofluoridate), a highly potent irreversible inhibitor of acetylcholinesterase (AChE), causes seizures and rapidly increases Fos and glial fibrillary acidic protein (GFAP) staining in piriform cortex (PC). pinacolymethylphosphonofluoridate 7-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-155 9570817-12 1998 Pretreatment with the selective muscarinic receptor antagonist scopolamine blocked the convulsions and prevented increased Fos and GFAP staining in PC. Scopolamine 63-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-126 9593956-0 1998 Pentylenetetrazole (PTZ)-induced c-fos expression in the hippocampus of kindled rats is suppressed by concomitant treatment with naloxone. Pentylenetetrazole 0-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 9593956-0 1998 Pentylenetetrazole (PTZ)-induced c-fos expression in the hippocampus of kindled rats is suppressed by concomitant treatment with naloxone. Pentylenetetrazole 20-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 9593956-0 1998 Pentylenetetrazole (PTZ)-induced c-fos expression in the hippocampus of kindled rats is suppressed by concomitant treatment with naloxone. Naloxone 129-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 9593956-5 1998 In the kindled animals, a pronounced but transient increase in c-fos mRNA level was observed in several brain areas after the injection of PTZ. Pentylenetetrazole 139-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 9593956-8 1998 The induction of c-fos expression in the hippocampus of stage five kindled rats but not in other brain areas was prevented by treatment of naloxone prior to each PTZ application. Naloxone 139-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 9593956-8 1998 The induction of c-fos expression in the hippocampus of stage five kindled rats but not in other brain areas was prevented by treatment of naloxone prior to each PTZ application. Pentylenetetrazole 162-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 9593956-10 1998 In addition, a single PTZ application (at the higher dose of 45 mg/kg) to rats that were not kindled also caused c-fos expression in several brain regions, but this was not influenced by naloxone. Pentylenetetrazole 22-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 9631428-0 1998 Seizures in rats treated with kainic acid induce Fos-like immunoreactivity in locus coeruleus. Kainic Acid 30-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 9631428-1 1998 Kainic acid-triggered seizures (KATS) induce Fos-like immunoreactivity (FLI) in limbic structures, which send efferents to the locus coeruleus (LC). Kainic Acid 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 9644036-1 1998 We have recently reported that an acute intragastric hypertonic saline load increases plasma arginine vasopressin (PAVP) and Fos immunoreactivity in several central nuclei, including the supraoptic nucleus (SON), paraventricular nucleus (PVN), nucleus of the solitary tract (NTS), area postrema (AP), and lateral parabrachial nucleus (LPBN). Sodium Chloride 64-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-128 9644036-3 1998 To test this hypothesis, we examined the effect of bilateral subdiaphragmatic vagotomy and bilateral splanchnic denervation on the PAVP and Fos immunoreactivity responses to intragastric hypertonic saline infusion in awake rats. Sodium Chloride 198-204 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-143 29090823-0 1998 The Serotonergic System Modulates the Cocaine-Induced Expression of the Immediate Early Genes egr-1 and c-fos in Rat Brain. Cocaine 38-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 29090823-6 1998 Cocaine-induced egr-1 and c-fos expression was substantially reduced in the brain areas from rats in which the serotonergic projections were lesioned by injection of the neurotoxin 5,7-dihydroxytryptamine, and in rats that have been injected with tropisetron, an antagonist of the 5-hydroxytryptamine (5-HT3) receptor. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 29090823-6 1998 Cocaine-induced egr-1 and c-fos expression was substantially reduced in the brain areas from rats in which the serotonergic projections were lesioned by injection of the neurotoxin 5,7-dihydroxytryptamine, and in rats that have been injected with tropisetron, an antagonist of the 5-hydroxytryptamine (5-HT3) receptor. 5,7-Dihydroxytryptamine 181-204 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 9602039-6 1998 Analysis of c-fos and c-jun expression at 2 h, by means of in situ hybridization, revealed diminished induction in dexamethasone-treated animals. Dexamethasone 115-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 9602069-6 1998 The blockade of KA-induced proENK and proDYN mRNA levels by the pre-treatment with L-ARG was well correlated with proto-oncoprotein levels, such as c-Fos, Fra-2, FosB, JunD, JunB, and c-Jun, as well as AP-1 and ENKCRE-2 DNA binding activities. Arginine 83-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-153 9602069-7 1998 The pre-administration with L-NAME further increased KA-induced c-jun and c-fos mRNA levels in addition to their protein product levels, although the pre-treatment with L-NAME did not affect KA-induced FosB, Fra-2, JunB, and JunD protein levels at 6 h after treatment. NG-Nitroarginine Methyl Ester 28-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 9602069-9 1998 Our results suggest that L-ARG plays an important role in inhibiting KA-induced proENK or proDYN mRNA expression, and its inhibitory action may be mediated through reducing the proto-oncoprotein levels, such as c-Fos, Fra-2, FosB, c-Jun, JunD, and JunB. Arginine 25-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-216 9602069-10 1998 In addition, L-NAME potentiated the c-Fos or c-Jun gene expression, as well as AP-1 or ENKCRE-2 DNA binding activity. NG-Nitroarginine Methyl Ester 13-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 9692675-3 1998 Interestingly, maximal c-fos-suppressing doses of D609 did not affect activity of extracellular signal-regulated kinases. tricyclodecane-9-yl-xanthogenate 50-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 10374349-8 1998 CONCLUSIONS: These data suggest that the stimulation of TRAb-IgG followed by cAMP production and 3H-thymidine incorporation is related to the induction of c-fos mRNA and, thus, to the growth of FRTL-5 cells. Cyclic AMP 77-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-160 10374349-8 1998 CONCLUSIONS: These data suggest that the stimulation of TRAb-IgG followed by cAMP production and 3H-thymidine incorporation is related to the induction of c-fos mRNA and, thus, to the growth of FRTL-5 cells. 3h-thymidine 97-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-160 9629957-1 1998 In rats with unilateral 6-hydroxydopamine (6-OHDA) lesion of the nigrostriatal pathway, amphetamine produces ipsiversive rotational behavior and activation of Fos in the intact striatum, but practically no activation of Fos in the denervated striatum. Amphetamine 88-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-162 9629957-1 1998 In rats with unilateral 6-hydroxydopamine (6-OHDA) lesion of the nigrostriatal pathway, amphetamine produces ipsiversive rotational behavior and activation of Fos in the intact striatum, but practically no activation of Fos in the denervated striatum. Amphetamine 88-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 220-223 9629957-4 1998 Injection of amphetamine (0.5 mg/kg or 5 mg/kg) induced contraversive rotation and strong and evenly distributed Fos expression in the lesioned striatum; in the contralateral striatum, however, Fos density was lower than in nonlesioned rats. Amphetamine 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-116 9629957-4 1998 Injection of amphetamine (0.5 mg/kg or 5 mg/kg) induced contraversive rotation and strong and evenly distributed Fos expression in the lesioned striatum; in the contralateral striatum, however, Fos density was lower than in nonlesioned rats. Amphetamine 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 194-197 9751140-4 1998 Administration of SKF 38393, a D1R receptor agonist, increased c-fos mRNA in ICjM, whereas administration of quinpirole, a D2R/D3R agonist, decreased it; SCH 23390, a D1 R antagonist and nafadotride, a preferential D3R antagonist, given alone, had effects opposite to those of the corresponding agonists. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 9751140-5 1998 These data indicate that basal c-fos expression in ICjM is maintained by endogenous dopamine acting tonically upon two receptor subtypes subserving opposite effects on the same cell. Dopamine 84-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 9751146-0 1998 Delta9-tetrahydrocannabinol increases sequence-specific AP-1 DNA-binding activity and Fos-related antigens in the rat brain. Dronabinol 0-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 9751146-3 1998 One hour after the intraperitoneal administration of delta9-THC at a dose of 10 or 15 mg/kg, AP-1 DNA-binding activity in the nucleus accumbens increased by 33 and 49%, respectively, while Western blot showed an increase in both c-Fos, FosB, Fra-1 (Fos-related antigen) and Fra-2. Dronabinol 53-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 229-234 9751146-6 1998 The effect of delta9-THC on the AP-1 DNA binding and the Fos-related antigens in the nucleus accumbens was blocked by the specific cannabinoid antagonist SR141716 A (3 mg/kg i.p.). Dronabinol 14-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 9751146-6 1998 The effect of delta9-THC on the AP-1 DNA binding and the Fos-related antigens in the nucleus accumbens was blocked by the specific cannabinoid antagonist SR141716 A (3 mg/kg i.p.). Cannabinoids 131-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 9751146-6 1998 The effect of delta9-THC on the AP-1 DNA binding and the Fos-related antigens in the nucleus accumbens was blocked by the specific cannabinoid antagonist SR141716 A (3 mg/kg i.p.). Rimonabant 154-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 9751146-8 1998 The results indicate that delta9-THC activates gene coding for AP-1 DNA-binding proteins by acting on cannabinoid receptors, and induces a different transcriptional program on the early-immediate gene of the Fos family, in different areas in the rat brain, suggesting that this mechanism might be involved in the central actions of cannabinoids. Dronabinol 26-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 208-211 9751146-8 1998 The results indicate that delta9-THC activates gene coding for AP-1 DNA-binding proteins by acting on cannabinoid receptors, and induces a different transcriptional program on the early-immediate gene of the Fos family, in different areas in the rat brain, suggesting that this mechanism might be involved in the central actions of cannabinoids. Cannabinoids 332-344 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 208-211 9593080-7 1998 Consistent with its effects on VSMC growth/proliferation, amlodipine also decreased c-myc, c-fos, and c-jun protooncogene expression induced by serum, thrombin, or bFGF within 1 h after cell activation, as assessed by semiquantitative reverse transcriptase (RT)-polymerase chain reaction (PCR) analysis. Amlodipine 58-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 9661997-0 1998 Effects of age on pilocarpine-induced c-fos expression in rat hippocampus and cortex. Pilocarpine 18-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 9661997-1 1998 The effect of age on pilocarpine-induced expression of the immediate-early gene c-fos was examined in the hippocampus and cortex of Long-Evans rats. Pilocarpine 21-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 9661997-5 1998 In saline-treated animals, comparable levels of c-fos mRNA and Fos-like protein were observed in the hippocampus and cortical regions of young (6 month) and aged (24-26 months) rats. Sodium Chloride 3-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 9661997-5 1998 In saline-treated animals, comparable levels of c-fos mRNA and Fos-like protein were observed in the hippocampus and cortical regions of young (6 month) and aged (24-26 months) rats. Sodium Chloride 3-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 9661997-6 1998 The expression of Fos-like protein following pilocarpine treatment was increased, however, in frontal, retrosplenial, and cingulate cortex of aged compared to young rats. Pilocarpine 45-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 9682220-0 1998 C-fos mediates cocaine inhibition of NGF-induced PC12 cell differentiation. Cocaine 15-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9682220-6 1998 Time course of c-fos expression up to 72 h was determined in cells treated with NGF 20 ng/ml and cocaine 10 microgram/ml (a moderately toxic level) by RT-PCR analysis. Cocaine 97-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 9682220-8 1998 In both control and experimental conditions, c-fos level was maximal at 0.5 h. In the control cells, c-fos expression declined rapidly to less than 5% of the 0.5h value, while in the cocaine-treated cells, c-fos level persisted through the 72-h exposure. Cocaine 183-190 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 9682220-8 1998 In both control and experimental conditions, c-fos level was maximal at 0.5 h. In the control cells, c-fos expression declined rapidly to less than 5% of the 0.5h value, while in the cocaine-treated cells, c-fos level persisted through the 72-h exposure. Cocaine 183-190 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 9682220-8 1998 In both control and experimental conditions, c-fos level was maximal at 0.5 h. In the control cells, c-fos expression declined rapidly to less than 5% of the 0.5h value, while in the cocaine-treated cells, c-fos level persisted through the 72-h exposure. Cocaine 183-190 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 9682220-9 1998 Adding c-fos antisense to cells treated with NGF and cocaine resulted in significant improvement of neurite out-growth, from 28% (NGF + cocaine) to 89% (NGF + cocaine + c-fos antisense) of control differentiation after 72 h of exposure (Dunnet"s T < 3.24). Cocaine 53-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 7-12 9682220-9 1998 Adding c-fos antisense to cells treated with NGF and cocaine resulted in significant improvement of neurite out-growth, from 28% (NGF + cocaine) to 89% (NGF + cocaine + c-fos antisense) of control differentiation after 72 h of exposure (Dunnet"s T < 3.24). Cocaine 53-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-174 9682220-9 1998 Adding c-fos antisense to cells treated with NGF and cocaine resulted in significant improvement of neurite out-growth, from 28% (NGF + cocaine) to 89% (NGF + cocaine + c-fos antisense) of control differentiation after 72 h of exposure (Dunnet"s T < 3.24). Cocaine 136-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 7-12 9682220-9 1998 Adding c-fos antisense to cells treated with NGF and cocaine resulted in significant improvement of neurite out-growth, from 28% (NGF + cocaine) to 89% (NGF + cocaine + c-fos antisense) of control differentiation after 72 h of exposure (Dunnet"s T < 3.24). Cocaine 136-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 7-12 9682220-10 1998 Inhibitory effects of cocaine on NGF-induced PC12 differentiation may be attributed to alteration of c-fos expression. Cocaine 22-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 9696479-0 1998 The NMDA receptor antagonist MK-801 reduces capsaicin-induced c-fos expression within rat trigeminal nucleus caudalis. Dizocilpine Maleate 29-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 9696479-0 1998 The NMDA receptor antagonist MK-801 reduces capsaicin-induced c-fos expression within rat trigeminal nucleus caudalis. Capsaicin 44-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 9696479-8 1998 30 min before capsaicin (5 nmol in 100 microl artificial CSF) reduced significantly and dose-dependently (12%, 36% and 47%, respectively) the c-fos-LI cells in TNC at each level from rostral to caudal but not in solitary tract, area postrema and lateral reticular nuclei, and for unexplained reasons, increased c-fos-LI within the inferior olive. Capsaicin 14-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-147 9552172-3 1998 cfos mRNA was induced in cortical areas at early times after either dose of PCP or of MK801; the change was especially prominent in cingulate and auditory cortices. Dizocilpine Maleate 86-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-4 10375734-5 1998 RESULTS: Exposure of cardiomyocytes to captopril (Cap, 100 mumol.L-1) for 48 h inhibited the rates of [3H]Urd and [3H]Leu incorporations by 25% and 26%, respectively, and for 2 h inhibited c-myc, c-fos mRNA expression by 75% and 55%, respectively. Captopril 39-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 196-201 10375734-5 1998 RESULTS: Exposure of cardiomyocytes to captopril (Cap, 100 mumol.L-1) for 48 h inhibited the rates of [3H]Urd and [3H]Leu incorporations by 25% and 26%, respectively, and for 2 h inhibited c-myc, c-fos mRNA expression by 75% and 55%, respectively. Captopril 50-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 196-201 10375734-6 1998 Treatment of angiotensin II (Ang II, 1 mumol.L-1) for 48 h significantly increased the rates of [3H]Urd and [3H]Leu incorporations and for 1 h induced c-myc, c-fos mRNA overexpression, which were reduced by pretreatment with Cap (100 mumol.L-1). Captopril 225-228 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 9593855-0 1998 Expression of Fos protein in the limbic regions of the rat following haloperidol decanoate. haloperidol decanoate 69-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 9593855-1 1998 To identify sites of antipsychotic drug action, the effects of acute and chronic haloperidol treatment on Fos protein expression in rat brain regions were examined by immunohistochemical methods. Haloperidol 81-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 9593855-5 1998 A single dose of haloperidol to chronic vehicle-treated rats produced significant increases in Fos-positive neurons in 18 of 21 brain regions examined including the several cortical areas, caudate-putamen, nucleus accumbens, lateral septum, thalamic nuclei, amygdala, hippocampus CA1, mesencephalic dopaminergic nuclei, and periaqueductal grey. Haloperidol 17-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 9593855-6 1998 The rats treated with acute vehicle after chronic haloperidol showed persistent Fos increases in confined brain regions comprising the lateral and central amygdala, lateral septum, and entorhinal cortex. Haloperidol 50-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 9593855-7 1998 Additional haloperidol injection to the chronic haloperidol-treated rats induced significant increases in Fos immunoreactivity in more widespread limbic-thalamo-cortical areas, whereas no significant increase was seen in the dorsolateral caudate-putamen. Haloperidol 11-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 9593855-7 1998 Additional haloperidol injection to the chronic haloperidol-treated rats induced significant increases in Fos immunoreactivity in more widespread limbic-thalamo-cortical areas, whereas no significant increase was seen in the dorsolateral caudate-putamen. Haloperidol 48-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 9753096-0 1998 Mapping of neuronal networks underlying generalized seizures induced by increasing doses of pentylenetetrazol in the immature and adult rat: a c-Fos immunohistochemical study. Pentylenetetrazole 92-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-148 9753096-7 1998 In P10 rats receiving a behaviourally non-active dose of PTZ, c-Fos immunoreactivity appeared only in the amygdala. Pentylenetetrazole 57-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 9753096-11 1998 In P21 and adult rats, the inactive dose of PTZ induced c-Fos immunoreactivity in thalamus and hypothalamus. Pentylenetetrazole 44-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 9607780-4 1998 The specific objectives were 1) to characterize the short term effects of BPA on cell proliferation and c-fos expression in the uterus and vagina, and 2) to compare the effects of prolonged exposure to low doses of BPA on the reproductive tract of F344 and SD rats. bisphenol A 74-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 9607780-7 1998 Quantitative RT-PCR revealed that both BPA and E2 increased c-fos messenger RNA levels in the uterus 14- to 16-fold within 2 h, which returned to basal levels after 6 h. In the vagina, BPA-induced c-fos expression remained elevated for up to 6 h, compared with the transient increase caused by E2. bisphenol A 39-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 9607780-7 1998 Quantitative RT-PCR revealed that both BPA and E2 increased c-fos messenger RNA levels in the uterus 14- to 16-fold within 2 h, which returned to basal levels after 6 h. In the vagina, BPA-induced c-fos expression remained elevated for up to 6 h, compared with the transient increase caused by E2. bisphenol A 39-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 197-202 9607780-7 1998 Quantitative RT-PCR revealed that both BPA and E2 increased c-fos messenger RNA levels in the uterus 14- to 16-fold within 2 h, which returned to basal levels after 6 h. In the vagina, BPA-induced c-fos expression remained elevated for up to 6 h, compared with the transient increase caused by E2. bisphenol A 185-188 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 9607780-7 1998 Quantitative RT-PCR revealed that both BPA and E2 increased c-fos messenger RNA levels in the uterus 14- to 16-fold within 2 h, which returned to basal levels after 6 h. In the vagina, BPA-induced c-fos expression remained elevated for up to 6 h, compared with the transient increase caused by E2. bisphenol A 185-188 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 197-202 9601697-1 1998 The 5HT1B agonist RU24969 (2.5-5.0 mg/kg) and anpirtoline (2.0 mg/kg) induced a striking increase in striatal Fos-like immunoreactivity in rats. 5-methoxy 3-(1,2,3,6-tetrahydro-4-pyridinyl)1H indole 18-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 9601697-1 1998 The 5HT1B agonist RU24969 (2.5-5.0 mg/kg) and anpirtoline (2.0 mg/kg) induced a striking increase in striatal Fos-like immunoreactivity in rats. anpirtoline 46-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 9548700-4 1998 Rats with constant corticosterone levels displayed enhanced footshock-induced Fos expression in the parvicellular compartment of the PVH, as well as in certain limbic and somatosensory cell groups, the locus coeruleus, but not in medullary catecholaminergic cell groups. Corticosterone 19-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 9526010-4 1998 Cell death caused by LY294002 resembles death caused by NGF deprivation in that it is blocked by a caspase inhibitor or a cAMP analog and that it is accompanied by the induction of c-jun, c-fos, and cyclin D1 mRNAs. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 21-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 188-193 9593045-1 1998 The fate of 15-mer phosphorothioate-modified antisense oligonucleotides to c-fos was followed after their microinjection into rat brain. Parathion 19-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 9593045-1 1998 The fate of 15-mer phosphorothioate-modified antisense oligonucleotides to c-fos was followed after their microinjection into rat brain. Oligonucleotides 55-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 9649921-0 1998 c-fos protein expression in apoptotic rat spermatocytes induced by gossypol. Gossypol 67-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9588485-4 1998 The AGm ablation reduced the numbers of amphetamine-stimulated Fos-immunoreactive nuclei in the ipsilateral dorsolateral striatum, where the AGm innervation is normally densest. Amphetamine 40-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 9582453-8 1998 This work, in conjunction with earlier work that demonstrated that cocaine and amphetamine have different effects on the expression of immediate early genes such as c-Fos, supports the hypothesis that these psychotropic agents evoke different patterns of gene expression which may lead to alteration in synaptic efficacy. Cocaine 67-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 167-170 9582453-8 1998 This work, in conjunction with earlier work that demonstrated that cocaine and amphetamine have different effects on the expression of immediate early genes such as c-Fos, supports the hypothesis that these psychotropic agents evoke different patterns of gene expression which may lead to alteration in synaptic efficacy. Amphetamine 79-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 167-170 9649921-2 1998 In this study, cellular levels of c-fos proteins in spermatocyte, either with or without gossypol exposure, were quantitatively detected by Western immunoblot and a computer-controlled Spot-denso-program with an IS-1000 Digital Imaging System. Gossypol 89-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 9649921-3 1998 Within 0.5-3.5 h (an average of 2 h) of the addition of gossypol, levels of c-fos proteins fell dramatically. Gossypol 56-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 9649921-4 1998 The reduction in c-fos proteins occurred 6 h before the apoptosis of spermatocytes in the presence of gossypol. Gossypol 102-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 9649921-5 1998 Four hours after exposure to gossypol, the c-fos protein content was overexpressed. Gossypol 29-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 9649921-8 1998 These results suggest that spermatocyte apoptosis induced by gossypol correlates with biphasic c-fos protein-mediated apoptosis. Gossypol 61-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 9566590-5 1998 Northern blot analysis showed that mRNA levels of c-fos, c-jun were elevated after treatment with ATP. Adenosine Triphosphate 98-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 9528974-7 1998 Quantitative solution hybridization analysis revealed a transient increase in c-fos mRNA levels by 20-fold following 30-45 min of LIF treatment, an effect that was inhibited by the tyrosine, as well as serine/threonine kinase inhibitors, genistein, and H7. Tyrosine 181-189 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 9528974-7 1998 Quantitative solution hybridization analysis revealed a transient increase in c-fos mRNA levels by 20-fold following 30-45 min of LIF treatment, an effect that was inhibited by the tyrosine, as well as serine/threonine kinase inhibitors, genistein, and H7. Genistein 238-247 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 9575042-8 1998 We demonstrate that detection of Fos in monkey brain tissue perfused with 4% paraformaldehyde can be improved by postfixation in a dilute acrolein solution. paraform 77-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 9575042-8 1998 We demonstrate that detection of Fos in monkey brain tissue perfused with 4% paraformaldehyde can be improved by postfixation in a dilute acrolein solution. Acrolein 138-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 9483552-6 1998 Fos immunoreactivity was marked in parvocellular regions of the paraventricular nucleus of the hypothalamus following both restraint periods and at both ages, an observation consistent with previous observations that restraint increases plasma corticosterone at both ages. Corticosterone 244-258 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9583764-0 1998 UP 202-56, an adenosine analogue, selectively acts via A1 receptors to significantly decrease noxiously-evoked spinal c-Fos protein expression. Adenosine 14-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 9678640-0 1998 Methamphetamine-induced Fos expression in the substantia nigra pars reticulata in rats with a unilateral 6-OHDA lesion of the nigrostriatal fibers. Methamphetamine 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 9678640-0 1998 Methamphetamine-induced Fos expression in the substantia nigra pars reticulata in rats with a unilateral 6-OHDA lesion of the nigrostriatal fibers. Oxidopamine 105-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 9583764-1 1998 The effects of oral administration of UP 202-56, an adenosine analogue, were assessed on carrageenan-induced spinal c-Fos protein expression and peripheral oedema. Carrageenan 89-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 9583764-2 1998 Three hours after intraplantar injection of carrageenan (6 mg/150 microl of saline), in awake rats, numerous c-Fos-like immunoreactive (c-Fos-LI) neurons in the dorsal horn of L4-L5 lumbar segments of the spinal cord (191 +/- 8; 184 +/- 10; 205 +/- 7 c-Fos-LI neurons per 40 microm section, for carrageenan controls in three experimental series performed in this study, respectively) and an extensive peripheral oedema were observed. Carrageenan 44-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 9583764-2 1998 Three hours after intraplantar injection of carrageenan (6 mg/150 microl of saline), in awake rats, numerous c-Fos-like immunoreactive (c-Fos-LI) neurons in the dorsal horn of L4-L5 lumbar segments of the spinal cord (191 +/- 8; 184 +/- 10; 205 +/- 7 c-Fos-LI neurons per 40 microm section, for carrageenan controls in three experimental series performed in this study, respectively) and an extensive peripheral oedema were observed. Carrageenan 44-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 9518601-2 1998 The present investigation sought to define brain regions affected by subanesthetic doses of ketamine, using high resolution autoradiographic analysis of 14C-2-deoxyglucose (2-DG) uptake and immunocytochemical staining for Fos-like immunoreactivity (Fos-LI). Ketamine 92-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 222-225 9583764-2 1998 Three hours after intraplantar injection of carrageenan (6 mg/150 microl of saline), in awake rats, numerous c-Fos-like immunoreactive (c-Fos-LI) neurons in the dorsal horn of L4-L5 lumbar segments of the spinal cord (191 +/- 8; 184 +/- 10; 205 +/- 7 c-Fos-LI neurons per 40 microm section, for carrageenan controls in three experimental series performed in this study, respectively) and an extensive peripheral oedema were observed. Carrageenan 44-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 9518601-2 1998 The present investigation sought to define brain regions affected by subanesthetic doses of ketamine, using high resolution autoradiographic analysis of 14C-2-deoxyglucose (2-DG) uptake and immunocytochemical staining for Fos-like immunoreactivity (Fos-LI). Ketamine 92-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 249-252 9583764-2 1998 Three hours after intraplantar injection of carrageenan (6 mg/150 microl of saline), in awake rats, numerous c-Fos-like immunoreactive (c-Fos-LI) neurons in the dorsal horn of L4-L5 lumbar segments of the spinal cord (191 +/- 8; 184 +/- 10; 205 +/- 7 c-Fos-LI neurons per 40 microm section, for carrageenan controls in three experimental series performed in this study, respectively) and an extensive peripheral oedema were observed. Sodium Chloride 76-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 9518601-7 1998 Ketamine induced Fos-LI in the same limbic cortical regions that exhibited increased 2-DG uptake in response to the subanesthetic dose of the drug. Ketamine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 9518601-7 1998 Ketamine induced Fos-LI in the same limbic cortical regions that exhibited increased 2-DG uptake in response to the subanesthetic dose of the drug. Deoxyglucose 85-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 9583764-2 1998 Three hours after intraplantar injection of carrageenan (6 mg/150 microl of saline), in awake rats, numerous c-Fos-like immunoreactive (c-Fos-LI) neurons in the dorsal horn of L4-L5 lumbar segments of the spinal cord (191 +/- 8; 184 +/- 10; 205 +/- 7 c-Fos-LI neurons per 40 microm section, for carrageenan controls in three experimental series performed in this study, respectively) and an extensive peripheral oedema were observed. Sodium Chloride 76-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 9518601-9 1998 Conversely, ketamine induced Fos in the paraventricular nucleus of the hypothalamus and central amygdala, although no effect of the drug on 2-DG uptake was apparent in these regions. Ketamine 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 9583764-2 1998 Three hours after intraplantar injection of carrageenan (6 mg/150 microl of saline), in awake rats, numerous c-Fos-like immunoreactive (c-Fos-LI) neurons in the dorsal horn of L4-L5 lumbar segments of the spinal cord (191 +/- 8; 184 +/- 10; 205 +/- 7 c-Fos-LI neurons per 40 microm section, for carrageenan controls in three experimental series performed in this study, respectively) and an extensive peripheral oedema were observed. Sodium Chloride 76-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 9518601-11 1998 By contrast, a robust induction of Fos-LI was observed after the anesthetic dose of ketamine that was neuroanatomically identical to that produced by the subanesthetic dose. Ketamine 84-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 9583764-3 1998 Oral UP 202-56 (10, 30 or 50 mg/kg) dose-dependently reduced the number of carrageenan-induced c-Fos-LI neurons (r = 0.931. Carrageenan 75-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 9518601-12 1998 Results of the present investigation show that anesthetic and subanesthetic doses of ketamine have pronounced effects on regional brain 2-DG uptake and induction of Fos-LI. Ketamine 85-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-168 9583764-4 1998 P < 0.0001), with the highest dose of UP 202-56 producing 72 +/- 4% reduction of the total number of carrageenan-induced spinal c-Fos-LI neurons, and 12 +/- 3% and 33 +/- 6% of reduction of control carrageenan oedema at paw and ankle levels, respectively. Carrageenan 104-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-136 9583764-7 1998 on the number of carrageenan-induced c-Fos-LI neurons. Carrageenan 17-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 9583764-11 1998 on both carrageenan-induced spinal c-Fos expression and peripheral oedema. Carrageenan 8-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 9518644-0 1998 Expression of c-fos and hsp70 mRNA in neonatal rat cerebrocortical slices during NMDA-induced necrosis and apoptosis. N-Methylaspartate 81-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 9518644-5 1998 After NMDA administration c-fos and hsp70 mRNA expression increased, with maxima occurring, respectively, at 1 h and 4 h after NMDA exposure. N-Methylaspartate 6-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 9518644-5 1998 After NMDA administration c-fos and hsp70 mRNA expression increased, with maxima occurring, respectively, at 1 h and 4 h after NMDA exposure. N-Methylaspartate 127-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 9518644-6 1998 When treatment with dizocilpine (MK-801; 10 microM), a non-competitive NMDA antagonist, was started before NMDA exposures, expression of both c-fos and hsp70 mRNA was decreased to values near control, indicating that activation of NMDA receptors induces both genes. Dizocilpine Maleate 20-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-147 9518644-6 1998 When treatment with dizocilpine (MK-801; 10 microM), a non-competitive NMDA antagonist, was started before NMDA exposures, expression of both c-fos and hsp70 mRNA was decreased to values near control, indicating that activation of NMDA receptors induces both genes. Dizocilpine Maleate 33-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-147 9518687-0 1998 Quinpirole attenuates the striatal fos expression induced by escape behavior. Quinpirole 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 9518687-1 1998 Several studies have shown that the D2-like dopamine receptor agonist quinpirole is able to markedly potentiate the striatal Fos expression induced by D1 agonists. Quinpirole 70-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-128 9518687-2 1998 The present study examined the effects of quinpirole on the striatal Fos-like immunoreactivity (FLI) induced by escape behavior. Quinpirole 42-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 9518687-6 1998 These findings demonstrate that quinpirole affects the striatal Fos expression induced by shuttling in a very different fashion than it does that induced by D1 agonists, and further support the view that dopaminergic mechanisms play an important role in behaviorally induced striatal Fos expression. Quinpirole 32-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 9518687-6 1998 These findings demonstrate that quinpirole affects the striatal Fos expression induced by shuttling in a very different fashion than it does that induced by D1 agonists, and further support the view that dopaminergic mechanisms play an important role in behaviorally induced striatal Fos expression. Quinpirole 32-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 284-287 9557971-0 1998 C-Fos-like immunoreactivity in the upper cervical spinal dorsal horn neurons following noxious chemical stimulation of the nasal mucosa in pentobarbital-anesthetized rats. Pentobarbital 139-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9557971-1 1998 Noxious chemical stimulation of the rat nasal mucosa with mustard oil induces the expression of Fos-like immunoreactivity in trigeminal and other brain stem neurons which contribute to upper airway protective reflexes such as sneezing, coughing and apnea. mustard oil 58-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-99 9557971-3 1998 Two hours after the application of mustard oil, numerous Fos-immunoreactive neurons were found in the mediolateral end of the C1 and dorsolateral division of the C2. mustard oil 35-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 9480923-0 1998 Exogenous C2-ceramide activates c-fos serum response element via Rac-dependent signalling pathway. N-acetylsphingosine 10-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 9480871-0 1998 Heparin inhibits Ca2+/calmodulin-dependent kinase II activation and c-fos induction in mesangial cells. Heparin 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 9480871-2 1998 In particular, heparin partially suppresses the ability of quiescent RMCs to enter the cell cycle and induce c-fos expression. Heparin 15-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 9480871-4 1998 However, we have also shown that heparin suppresses c-fos expression in response to ionophores such as ionomycin, an event independent of mitogen-activated protein kinase [Miralem, Wang, Whiteside and Templeton (1996) J. Biol. Heparin 33-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 9480871-4 1998 However, we have also shown that heparin suppresses c-fos expression in response to ionophores such as ionomycin, an event independent of mitogen-activated protein kinase [Miralem, Wang, Whiteside and Templeton (1996) J. Biol. Ionomycin 103-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 9480871-10 1998 The broad-specificity CaMK inhibitor, KT 5926, inhibited ionomycin-dependent c-fos induction at a concentration at which it was without effect on induction by serum or phorbol ester. KT 5926 38-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 9480871-10 1998 The broad-specificity CaMK inhibitor, KT 5926, inhibited ionomycin-dependent c-fos induction at a concentration at which it was without effect on induction by serum or phorbol ester. Ionomycin 57-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 9480871-11 1998 The CaMK II-specific inhibitor, KN-93, likewise inhibited c-fos induction by ionomycin, but not by serum or phorbol ester. KN 93 32-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 9480871-11 1998 The CaMK II-specific inhibitor, KN-93, likewise inhibited c-fos induction by ionomycin, but not by serum or phorbol ester. Ionomycin 77-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 9480871-13 1998 Heparin (1 microg/ml) suppressed ionomycin-dependent c-fos induction. Heparin 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 9480871-13 1998 Heparin (1 microg/ml) suppressed ionomycin-dependent c-fos induction. Ionomycin 33-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 9480871-18 1998 We conclude that ionomycin induces c-fos in RMCs through the CaMK II pathway, and that heparin prevents CaMK II activation by an indirect process mediated by other cell components. Ionomycin 17-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 9555037-3 1998 In the present study, involvement of Fos in the mechanism of the PENK and PDYN gene induction in the hippocampal dentate gyrus during seizures elicited by kainic acid was studied using a knock-down technique. Kainic Acid 155-166 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 9673793-0 1998 Pattern of c-fos mRNA induction in rat brain by acute morphine. Morphine 54-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 9673793-4 1998 This study was designed to identify brain regions that demonstrate specific induction of the IEG c-fos, a component of the AP-1 transcription factor, in response to acute morphine, and to contrast this induction with the stressful effects of the injection itself. Morphine 171-179 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 9673793-7 1998 Specific induction of c-fos mRNA by morphine was seen in dorsomedial caudate-putamen, paraventricular nucleus of the thalamus, central and intralaminar thalamic nuclei, dorsal central grey, superior colliculus, lateral parabrachial nucleus, inferior olivary complex, and caudal nucleus tractus solitarius. Morphine 36-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 9673793-8 1998 These findings represent the first complete anatomical mapping of c-fos induction in rat brain, and show that acute morphine administration alters gene expression in several areas related to known functional properties of opioids. Morphine 116-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 9555073-4 1998 In a separate experiment, intrastriatal DNQX was shown to block kainic acid (KA)-induced Fos expression in the striatum, but not in adjacent cerebral cortex, suggesting that the diffusion of this drug is restricted to the striatum. FG 9041 40-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 9555073-4 1998 In a separate experiment, intrastriatal DNQX was shown to block kainic acid (KA)-induced Fos expression in the striatum, but not in adjacent cerebral cortex, suggesting that the diffusion of this drug is restricted to the striatum. Kainic Acid 64-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 9555087-0 1998 Noxious heat-evoked fos-like immunoreactivity in the rat lumbar dorsal horn is inhibited by glutamate microinjections in the upper cervical spinal cord. Glutamic Acid 92-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 9555087-1 1998 Microinjections of glutamate into the upper cervical spinal cord significantly reduced (to 57% of control) the total number of neurons demonstrating noxious heat-evoked fos-like immunoreactivity in the lumbar spinal cord. Glutamic Acid 19-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-172 9555037-6 1998 The subsequent induction of PENK and PDYN mRNAs was reduced by more than 60% by the c-fos antisense oligonucleotide, while constitutive expression of three other genes (alpha-tubulin, NMDA receptor-1, and GS protein alpha-subunit) was not affected. Oligonucleotides 100-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 9480923-2 1998 To understand the signal transduction pathway of ceramide to the nucleus, in the present study, we examined whether C2-ceramide, a cell permeable ceramide, activates c-fos serum response element (SRE). N-acetylsphingosine 116-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-171 9480923-2 1998 To understand the signal transduction pathway of ceramide to the nucleus, in the present study, we examined whether C2-ceramide, a cell permeable ceramide, activates c-fos serum response element (SRE). Ceramides 119-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-171 9480923-3 1998 Treatment of Rat-2 fibroblast cells with C2-ceramide caused the stimulation of c-fos SRE-dependent reporter gene activity in a dose- and time-dependent manner by transient transfection analysis. N-acetylsphingosine 41-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 9501868-16 1998 The induction of c-fos and c-jun gene expression by bFGF or by PMA was blocked by the PKC inhibitors H-7 (30 microM) or GF109203X (1 microM). 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine 101-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 9501868-16 1998 The induction of c-fos and c-jun gene expression by bFGF or by PMA was blocked by the PKC inhibitors H-7 (30 microM) or GF109203X (1 microM). bisindolylmaleimide I 120-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 9625354-0 1998 Separate populations of neurons within the paraventricular hypothalamic nucleus of the rat project to vagal and thoracic autonomic preganglionic levels and express c-Fos protein induced by lithium chloride. Lithium Chloride 189-205 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-169 9659456-1 1998 OBJECTIVES: The aims of this study were to determine (1) whether neonatal rat cardiac fibroblasts (CAFB) express P2Y receptors; (2) whether CAFB respond to extracellular ATP by inducing expression of c-fos mRNA; and (3) whether extracellular ATP modulates norepinephrine (NE)-stimulated cell growth in CAFB. Adenosine Triphosphate 170-173 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 200-205 9659456-1 1998 OBJECTIVES: The aims of this study were to determine (1) whether neonatal rat cardiac fibroblasts (CAFB) express P2Y receptors; (2) whether CAFB respond to extracellular ATP by inducing expression of c-fos mRNA; and (3) whether extracellular ATP modulates norepinephrine (NE)-stimulated cell growth in CAFB. Adenosine Triphosphate 242-245 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 200-205 9659456-1 1998 OBJECTIVES: The aims of this study were to determine (1) whether neonatal rat cardiac fibroblasts (CAFB) express P2Y receptors; (2) whether CAFB respond to extracellular ATP by inducing expression of c-fos mRNA; and (3) whether extracellular ATP modulates norepinephrine (NE)-stimulated cell growth in CAFB. Norepinephrine 256-270 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 200-205 9659456-1 1998 OBJECTIVES: The aims of this study were to determine (1) whether neonatal rat cardiac fibroblasts (CAFB) express P2Y receptors; (2) whether CAFB respond to extracellular ATP by inducing expression of c-fos mRNA; and (3) whether extracellular ATP modulates norepinephrine (NE)-stimulated cell growth in CAFB. cafb 140-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 200-205 9659456-6 1998 Extracellular ATP induced the expression of c-fos mRNA through a pathway that was sensitive to inhibitors of protein kinase C (PKC), but not to inhibitors of intracellular Ca2+ signaling. Adenosine Triphosphate 14-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 9659456-8 1998 Whereas the potency order for stimulation of c-fos expression was ATP = UTP > ADP > adenosine, the potency order to inhibit the NE-induced increase of [3H]thymidine incorporation into DNA was ATP > ADP > UTP > adenosine. Adenosine Triphosphate 66-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 9659456-8 1998 Whereas the potency order for stimulation of c-fos expression was ATP = UTP > ADP > adenosine, the potency order to inhibit the NE-induced increase of [3H]thymidine incorporation into DNA was ATP > ADP > UTP > adenosine. Uridine Triphosphate 72-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 9659456-8 1998 Whereas the potency order for stimulation of c-fos expression was ATP = UTP > ADP > adenosine, the potency order to inhibit the NE-induced increase of [3H]thymidine incorporation into DNA was ATP > ADP > UTP > adenosine. Adenosine Diphosphate 81-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 9659456-9 1998 CONCLUSIONS: These data demonstrate that CAFB express both P2Y1 and P2Y2 receptor mRNA and that CAFB respond to P2Y receptor stimulation by induction of c-fos and inhibition of DNA synthesis. cafb 96-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-158 9515031-3 1998 In the transient transfection assay, TGF-beta1 potentiated NE- or 12-O-tetradecanoylphorbol-13-acetate (TPA)-activated c-fos promoter/enhancer, but not forskolin-activated c-fos promoter/enhancer. Tetradecanoylphorbol Acetate 66-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 9515031-3 1998 In the transient transfection assay, TGF-beta1 potentiated NE- or 12-O-tetradecanoylphorbol-13-acetate (TPA)-activated c-fos promoter/enhancer, but not forskolin-activated c-fos promoter/enhancer. Tetradecanoylphorbol Acetate 104-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 9515031-3 1998 In the transient transfection assay, TGF-beta1 potentiated NE- or 12-O-tetradecanoylphorbol-13-acetate (TPA)-activated c-fos promoter/enhancer, but not forskolin-activated c-fos promoter/enhancer. Tetradecanoylphorbol Acetate 104-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-177 9515031-4 1998 The c-fos serum response element (SRE) and the TPA response element (TRE) were responsible for TGF-beta1-induced potentiation of the NE or TPA action. Tetradecanoylphorbol Acetate 139-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 9515031-5 1998 Although TGF-beta1 activated not only the wild-type c-fos SRE, but also the mutated c-fos SRE, which contains an intact binding site for the serum response factor (SRF) but lacks the ternary complex factor (TCF) binding site, TPA activated the wild-type c-fos SRE but not the mutated c-fos SRE. Tetradecanoylphorbol Acetate 226-229 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 9515031-5 1998 Although TGF-beta1 activated not only the wild-type c-fos SRE, but also the mutated c-fos SRE, which contains an intact binding site for the serum response factor (SRF) but lacks the ternary complex factor (TCF) binding site, TPA activated the wild-type c-fos SRE but not the mutated c-fos SRE. Tetradecanoylphorbol Acetate 226-229 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 9515031-5 1998 Although TGF-beta1 activated not only the wild-type c-fos SRE, but also the mutated c-fos SRE, which contains an intact binding site for the serum response factor (SRF) but lacks the ternary complex factor (TCF) binding site, TPA activated the wild-type c-fos SRE but not the mutated c-fos SRE. Tetradecanoylphorbol Acetate 226-229 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 9465005-4 1998 It was also examined in lesioned rats whether the grafts may regulate the expression of c-Fos after systemic administration of apomorphine in the basal ganglia nuclei as well as their target structures, including the ventromedial thalamic nucleus (VM), superior colliculus (SC), and pedunculopontine nucleus (PPN). Apomorphine 127-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 9465005-7 1998 Apomorphine induced an increased expression of c-Fos in the GP, STN, VM, SC, and PPN on the lesioned side. Apomorphine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 9625354-8 1998 LiCl was used as stimulus for c-Fos-like immunohistochemistry. Lithium Chloride 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 9625354-10 1998 A significant number of FG/Fos double-labelled neurons were located in the dorsal parvocellular subnucleus of the PVN (dp) in the LiCl-stimulated rats. Lithium Chloride 130-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 9625354-12 1998 The presence of FG/Fos double-labelled neurons in the dp suggests that this nucleus could mediate a sympathetic response after LiCl administration. dp 54-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 9625354-12 1998 The presence of FG/Fos double-labelled neurons in the dp suggests that this nucleus could mediate a sympathetic response after LiCl administration. Lithium Chloride 127-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 9460760-5 1998 Combined pretreatment with CP99,994 and the competitive N-methyl-D-aspartate receptor antagonist, CPP, caused a greater reduction in c-fos expression at the subnucleus caudalis/cervical cord transition than after either drug alone suggesting interaction between receptors for glutamate and substance P. Tachykinin receptor antagonists did not reduce the number of Fos-positive neurons produced at the subnucleus interpolaris/subnucleus caudalis transition. cp99 27-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 364-367 9460759-2 1998 The present study examined the staining of c-fos-like immunoreactivity following opiate withdrawal or swim-stress (2.5-3 min at 21 degrees C) in periaqueductal gray neurons of the rat which had projections to and through the rostral ventromedial medulla identified by microinjection of the retrograde tracer, Fast Blue, into the nucleus raphe magnus prior to development of morphine dependence. Morphine 374-382 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 9460759-3 1998 Both naloxone-precipitated withdrawal and swim-stress increased numbers of neurons expressing c-fos-like immunoreactivity in periaqueductal gray. Naloxone 5-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 9460760-3 1998 The number of Fos-positive neurons produced in superficial laminae at the trigeminal subnucleus caudalis/cervical cord transition by application of the selective small fiber excitant, mustard oil, to the corneal surface was reduced by the neurokinin 1 receptor antagonist, CP99,994 (5-100 nmol, i.c.v.) mustard oil 184-195 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 9466401-0 1998 Mediation by N-methyl-D-aspartate and non-N-methyl-D-aspartate receptors in the expression of Fos protein at the nucleus tractus solitarii in response to baroreceptor activation in the rat. N-Methylaspartate 13-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 9466401-7 1998 The expression of Fos protein in this fashion was reduced, simultaneous with a discernible depression in baroreceptor reflex response, when baroreceptor activation was coupled with microinjection bilaterally of dizocilpine maleate (200 pmol) or 6-cyano-7-nitroquinoxaline-2,3-dione (8 pmol) into the nucleus tractus solitarii. Dizocilpine Maleate 211-230 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 9466401-7 1998 The expression of Fos protein in this fashion was reduced, simultaneous with a discernible depression in baroreceptor reflex response, when baroreceptor activation was coupled with microinjection bilaterally of dizocilpine maleate (200 pmol) or 6-cyano-7-nitroquinoxaline-2,3-dione (8 pmol) into the nucleus tractus solitarii. 6-Cyano-7-nitroquinoxaline-2,3-dione 245-281 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 9460760-3 1998 The number of Fos-positive neurons produced in superficial laminae at the trigeminal subnucleus caudalis/cervical cord transition by application of the selective small fiber excitant, mustard oil, to the corneal surface was reduced by the neurokinin 1 receptor antagonist, CP99,994 (5-100 nmol, i.c.v.) cp99 273-277 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 9460760-5 1998 Combined pretreatment with CP99,994 and the competitive N-methyl-D-aspartate receptor antagonist, CPP, caused a greater reduction in c-fos expression at the subnucleus caudalis/cervical cord transition than after either drug alone suggesting interaction between receptors for glutamate and substance P. Tachykinin receptor antagonists did not reduce the number of Fos-positive neurons produced at the subnucleus interpolaris/subnucleus caudalis transition. cp99 27-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 9466401-11 1998 These results suggest that glutamatergic neurotransmission plays an active role, via comparable contributions from both N-methyl-D-aspartate and non-N-methyl-D-aspartate receptors, in the expression of Fos protein at the caudal nucleus tractus solitarii in response to baroreceptor activation. N-Methylaspartate 120-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 202-205 9518565-4 1998 Dizocilpine also suppressed c-fos expression induced by dehydration in the median preoptic nucleus (MPN), the supraoptic and paraventricular nuclei (SON and PVN), but did not influence c-fos expression in the subfornical organ (SFO). Dizocilpine Maleate 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 9518565-6 1998 Double staining showed that suppression of c-fos expression following dizocilpine occurred in the NMDA R1 receptor containing neurons in the hypothalamus. Dizocilpine Maleate 70-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 9518565-8 1998 They complement our earlier findings that dizocilpine also attenuates drinking and c-fos expression following intraventricular infusions of angiotensin II. Dizocilpine Maleate 42-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 9500968-0 1998 Laminar distribution of Fos/calcium-binding protein and Fos/neurofilament protein-labeled neurons in rat motor and sensory cortex after picrotoxin-induced seizures. Picrotoxin 136-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 9530928-0 1998 Sexually dimorphic MK801-induced c-fos in the rat hypothalamic paraventricular nucleus. Dizocilpine Maleate 19-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 9517827-0 1998 Learned tolerance to ethanol-induced c-Fos expression in rats. Ethanol 21-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 9517827-1 1998 With c-Fos immunoreactivity as a marker for neural activity, we examined whether environmental cues associated with ethanol injection influence the expression of tolerance to ethanol-induced c-Fos activation. Ethanol 175-182 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 191-196 9517827-3 1998 Relative to rats that received ethanol for the first time, ethanol-induced c-Fos expression in the paraventricular nucleus of the hypothalamus (PVN) and the locus coeruleus (LC) was significantly reduced in rats that had received multiple prior ethanol administrations. Ethanol 31-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 9517827-3 1998 Relative to rats that received ethanol for the first time, ethanol-induced c-Fos expression in the paraventricular nucleus of the hypothalamus (PVN) and the locus coeruleus (LC) was significantly reduced in rats that had received multiple prior ethanol administrations. Ethanol 59-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 9517827-3 1998 Relative to rats that received ethanol for the first time, ethanol-induced c-Fos expression in the paraventricular nucleus of the hypothalamus (PVN) and the locus coeruleus (LC) was significantly reduced in rats that had received multiple prior ethanol administrations. Ethanol 59-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 9517827-5 1998 Results suggest that tolerance to ethanol, as indexed by c-Fos expression in the PVN and the LC, is mediated in part by Pavlovian conditioned responses to cues that predict ethanol administration. Ethanol 34-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 9517827-5 1998 Results suggest that tolerance to ethanol, as indexed by c-Fos expression in the PVN and the LC, is mediated in part by Pavlovian conditioned responses to cues that predict ethanol administration. Ethanol 173-180 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 9502431-6 1998 Similarly, expression of c-fos antisense RNA reduced by 80% the stimulatory effects of PACAP. pacap 87-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 9571013-0 1998 Testosterone augments neuronal Fos responses to estrous odors throughout the vomeronasal projection pathway of gonadectomized male and female rats. Testosterone 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 9571013-1 1998 Pheromonal signals emanating from female rats" soiled bedding have previously been shown to attract male conspecifics and to augment the number of Fos-immunoreactive neurons present in portions of the vomeronasal projection pathway, ranging from the accessory olfactory bulb (AOB) to the medial preoptic area (mPOA) of gonadectomized, testosterone-treated male as well as female subjects. Testosterone 335-347 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-150 9571013-2 1998 In the present study we extended these findings by showing that these neuronal Fos responses to estrous odors occurred only in gonadectomized subjects which received testosterone propionate (TP), as opposed to oil vehicle, at the time of testing. Testosterone Propionate 166-189 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 9571013-2 1998 In the present study we extended these findings by showing that these neuronal Fos responses to estrous odors occurred only in gonadectomized subjects which received testosterone propionate (TP), as opposed to oil vehicle, at the time of testing. Testosterone Propionate 191-193 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 9571013-5 1998 We propose that testosterone facilitates odor-induced neuronal Fos expression either via its conversion to estradiol, and the subsequent action of this steroid at estrogen response elements on the c-fos gene, or via some indirect mechanism involving centrifugal control of AOB neurotransmission. Testosterone 16-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 9571013-5 1998 We propose that testosterone facilitates odor-induced neuronal Fos expression either via its conversion to estradiol, and the subsequent action of this steroid at estrogen response elements on the c-fos gene, or via some indirect mechanism involving centrifugal control of AOB neurotransmission. Testosterone 16-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 197-202 9571013-5 1998 We propose that testosterone facilitates odor-induced neuronal Fos expression either via its conversion to estradiol, and the subsequent action of this steroid at estrogen response elements on the c-fos gene, or via some indirect mechanism involving centrifugal control of AOB neurotransmission. Estradiol 107-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 9571013-5 1998 We propose that testosterone facilitates odor-induced neuronal Fos expression either via its conversion to estradiol, and the subsequent action of this steroid at estrogen response elements on the c-fos gene, or via some indirect mechanism involving centrifugal control of AOB neurotransmission. Steroids 152-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 197-202 9488502-8 1998 Expression of the proto-oncogenes c-fos and c-jun was enhanced in ATP-treated animals as compared with controls. Adenosine Triphosphate 66-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 9500968-1 1998 Cerebrocortical Fos induction after picrotoxin-induced seizure occurs in spiny neurons and, to a lesser extent, in neurons defined by calcium-binding protein immunoreactivity. Picrotoxin 36-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 9500968-1 1998 Cerebrocortical Fos induction after picrotoxin-induced seizure occurs in spiny neurons and, to a lesser extent, in neurons defined by calcium-binding protein immunoreactivity. Calcium 134-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 9447994-1 1998 Several studies have characterized the upstream regulatory region of c-fos, and identified cis-acting elements termed the cyclic AMP (cAMP) response elements (CREs) that are critical for c-fos transcription in response to a variety of extracellular stimuli. Cyclic AMP 122-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 187-192 9649293-4 1998 Desensitization was studied by comparing the number of Fos-IR cells induced by 1% acetic acid in rats treated 24 hours before with 1 mM. Acetic Acid 82-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 9649293-6 1998 RESULTS: Lidocaine instilled previously markedly reduced the number of Fos-IR spinal cells responding to capsaicin-induced bladder afferent excitation. Lidocaine 9-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 9649293-6 1998 RESULTS: Lidocaine instilled previously markedly reduced the number of Fos-IR spinal cells responding to capsaicin-induced bladder afferent excitation. Capsaicin 105-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 9649293-7 1998 Numbers of Fos-IR cells induced by acetic acid instillation in bladders desensitized by capsaicin administrated 24 hours before were not changed by lidocaine application prior to capsaicin. Acetic Acid 35-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 9649293-7 1998 Numbers of Fos-IR cells induced by acetic acid instillation in bladders desensitized by capsaicin administrated 24 hours before were not changed by lidocaine application prior to capsaicin. Capsaicin 88-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 9447994-9 1998 Moreover, glutamate induction of c-fos expression in primary cortical neurons was dependent on CREB. Glutamic Acid 10-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 9453550-5 1998 Consistent with this cytoplasmic localization, inhibition of ECT-induced p42/p44MAPK activation by the extracellular signal-regulated kinase kinase inhibitor PD098059 blocked phosphorylation of the cytoplasmic protein microtubule-associated protein 2c (MAP2c), but failed to inhibit the induction of the nuclear protein c-Fos in response to ECT. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 158-166 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 320-325 9453550-7 1998 Accordingly, PD098059 blocked the induction of Fos-like immunoreactivity in the nuclei of cortical neurons as well as MAP2c phosphorylation in the cytoplasm. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 9466425-3 1998 Treatment with dibutyryl cyclic AMP, a permeable cyclic AMP, increased GABA uptake and immunocytochemically detectable levels of proteins such as c-Fos and calbindin-D28k. Bucladesine 15-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-151 9466425-3 1998 Treatment with dibutyryl cyclic AMP, a permeable cyclic AMP, increased GABA uptake and immunocytochemically detectable levels of proteins such as c-Fos and calbindin-D28k. Cyclic AMP 25-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-151 9447994-1 1998 Several studies have characterized the upstream regulatory region of c-fos, and identified cis-acting elements termed the cyclic AMP (cAMP) response elements (CREs) that are critical for c-fos transcription in response to a variety of extracellular stimuli. Cyclic AMP 134-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 187-192 9450515-2 1998 Fos expression in this region could also be induced by injections of the D2-like dopamine antagonist raclopride (0.5 mg/kg). Dopamine 81-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9574865-0 1998 A possible role for protein kinase C in CO2/H+-induced c-fos mRNA expression in PC12 cells. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 40-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 9574865-1 1998 Recently we have found that hypercapnia induces nuclear protein (FOS) expression in the brainstem chemosensitive neurons, including catecholamine-containing cells. Catecholamines 132-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 9574865-2 1998 In the present studies we examined the role of protein kinase C (PKC) pathway in CO2-induced c-fos expression. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 81-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 9574865-6 1998 Depletion of PKC abolished the effect of CO2 on c-fos mRNA expression, inhibited MAP kinases tyrosine phosphorylation and suppressed the expression of PKC(alpha) and PKC(zeta). N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 41-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 9574865-7 1998 These results suggest that MAP kinases, PKC(alpha) and/or PKC(beta) might be involved in CO2-induced c-fos mRNA expression. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 89-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 9450515-2 1998 Fos expression in this region could also be induced by injections of the D2-like dopamine antagonist raclopride (0.5 mg/kg). Raclopride 101-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9450515-4 1998 SCH-23390 was also able to prevent the atypical neuroleptic clozapine (30 mg/kg) from inducing Fos expression in the islands of Calleja. SCH 23390 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 9450515-4 1998 SCH-23390 was also able to prevent the atypical neuroleptic clozapine (30 mg/kg) from inducing Fos expression in the islands of Calleja. Clozapine 60-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 9531466-3 1998 Replacement of the calcium in the bathing medium by 2 mM manganese suppressed all detectable c-fos-ir, whereas inclusion of 0.5 microM capsaicin caused intense c-fos-ir expression in the absence of stimulation. Calcium 19-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 9531466-3 1998 Replacement of the calcium in the bathing medium by 2 mM manganese suppressed all detectable c-fos-ir, whereas inclusion of 0.5 microM capsaicin caused intense c-fos-ir expression in the absence of stimulation. Manganese 57-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 9531466-3 1998 Replacement of the calcium in the bathing medium by 2 mM manganese suppressed all detectable c-fos-ir, whereas inclusion of 0.5 microM capsaicin caused intense c-fos-ir expression in the absence of stimulation. Capsaicin 135-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 9531466-6 1998 Cords obtained from animals treated at 1 day old with capsaicin to destroy afferent C fibres showed a reduction in the number of c-fos-ir positive cells induced by high intensity dorsal root stimulation. Capsaicin 54-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 9519246-8 1998 With the same captopril pretreatment, both Fos- and Egr-1-ir in the SFO, MnPO, SON, PVN, AP and LPBN were also significantly reduced. Captopril 14-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 9548387-0 1998 MK-801 reverses Fos expression induced by the full dopamine D1 receptor agonist SKF-82958 in the rat striatum. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 9548387-0 1998 MK-801 reverses Fos expression induced by the full dopamine D1 receptor agonist SKF-82958 in the rat striatum. SK and F 82958 80-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 9548387-3 1998 The 3 mg/kg SKF-82958-induced expression of striatal Fos protein was blocked by the dopamine D1 receptor antagonist SCH-23390 (R(+)-7-chloro-8-hydroxy-3-methyl-1-phenyl-2,3,4,5-tetrahydro-1H-3-benza zepine) (0.3 mg/kg i.p.). SK and F 82958 12-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 9548387-3 1998 The 3 mg/kg SKF-82958-induced expression of striatal Fos protein was blocked by the dopamine D1 receptor antagonist SCH-23390 (R(+)-7-chloro-8-hydroxy-3-methyl-1-phenyl-2,3,4,5-tetrahydro-1H-3-benza zepine) (0.3 mg/kg i.p.). SCH 23390 116-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 9548387-3 1998 The 3 mg/kg SKF-82958-induced expression of striatal Fos protein was blocked by the dopamine D1 receptor antagonist SCH-23390 (R(+)-7-chloro-8-hydroxy-3-methyl-1-phenyl-2,3,4,5-tetrahydro-1H-3-benza zepine) (0.3 mg/kg i.p.). r(+)-7-chloro-8-hydroxy-3-methyl-1-phenyl-2,3,4,5-tetrahydro-1h-3-benza zepine 127-205 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 9548387-5 1998 also completely prevented striatal Fos induction by an injection of 3 mg/kg SKF-82958. 1,2,3,4-tetrahydroisoquinoline-7-sulfonamide 76-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 9507971-2 1998 The glucose antimetabolite, 2-deoxy-D-glucose (2DG), elicits expression of the proto-oncogene product Fos, which is expressed in hypothalamic structures where DA is synthesized. Glucose 4-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 9507971-2 1998 The glucose antimetabolite, 2-deoxy-D-glucose (2DG), elicits expression of the proto-oncogene product Fos, which is expressed in hypothalamic structures where DA is synthesized. Deoxyglucose 28-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 9507971-2 1998 The glucose antimetabolite, 2-deoxy-D-glucose (2DG), elicits expression of the proto-oncogene product Fos, which is expressed in hypothalamic structures where DA is synthesized. Deoxyglucose 47-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 9507971-2 1998 The glucose antimetabolite, 2-deoxy-D-glucose (2DG), elicits expression of the proto-oncogene product Fos, which is expressed in hypothalamic structures where DA is synthesized. Dopamine 159-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 9507971-6 1998 Whereas Fos-ir was negligible after saline administration, 2DG induced expression of Fos-ir by TH-ir neurons in the paraventricular (PVN), periventricular (Pe) and arcuate nuclei (ARC), and in the anterior hypothalamic area (AHA). Deoxyglucose 59-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 9507971-9 1998 These results reveal the functional responsiveness of discrete DA neuron populations to glucoprivation, and indicate that estradiol enhances cellular accumulation of Fos-ir by ARC DA neurons during this metabolic challenge. Estradiol 122-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-169 9507929-0 1998 Differential patterns of regional c-Fos induction in the rat brain by amphetamine and the novel wakefulness-promoting agent modafinil. Amphetamine 70-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 9507929-0 1998 Differential patterns of regional c-Fos induction in the rat brain by amphetamine and the novel wakefulness-promoting agent modafinil. Modafinil 124-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 9507929-2 1998 Both modafinil and amphetamine induced neuronal expression of c-Fos-like immunoreactivity in the paraventricular nucleus of the hypothalamus, anterior hypothalamus and central nucleus of the amygdala. Modafinil 5-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 9507929-2 1998 Both modafinil and amphetamine induced neuronal expression of c-Fos-like immunoreactivity in the paraventricular nucleus of the hypothalamus, anterior hypothalamus and central nucleus of the amygdala. Amphetamine 19-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 9507929-3 1998 Modafinil also increased c-Fos-like immunoreactivity in the suprachiasmatic nucleus, while amphetamine had no effect. Modafinil 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 9507929-4 1998 Brain regions in which amphetamine increased c-Fos-like immunoreactivity, but modafinil had no effect, included frontal cortex, striatum, lateral habenula, supraoptic nucleus and basolateral nucleus of the amygdala. Amphetamine 23-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 9548402-1 1998 Role of protein kinase C. Noradrenaline increased the mRNA levels of c-fos and c-jun in rat-1 fibroblast lines stably expressing the cloned alpha1-adrenoceptor subtypes. Norepinephrine 26-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 9502224-2 1998 The first part of the experiments showed that when formalin was injected into one hindpaw, the nociceptive c-fos expression in the lumbar dorsal horn ipsilateral to the injection was suppressed dose-dependently by intrathecal (i.t.) Formaldehyde 51-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 9502224-4 1998 In the second part of the study, the formalin injection was carried out into two hindpaws of the rats with a sectioned dorsal quadrant at the thoracic spinal level, in these rats, there was a significant suppression of c-fos expression in the dorsal horn on the side with intact dorsal quadrant, reasonably owing to the preservation of the spinally descending inhibitory fibers from the supraspinal level; furthermore, this suppression could be canceled following i.t. Formaldehyde 37-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 219-224 9473576-3 1998 Following inhalation of 3 and 7% CO2 in air for 5 min, the density of c-Fos-immunoreactive (IR) neurons increased stepwise not only in the 3rd-5th divisions of the VMS (between the caudal end of the nucleus corporis trapezoidei and the caudal end of the area postrema), but also in the rostroventromedial medulla (RVMM). N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 33-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 9507050-0 1998 Methyl palmoxirate increases eating behavior and brain Fos-like immunoreactivity in rats. methyl 2-tetradecylglycidate 0-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 9447868-0 1998 Comparison of the effects of treatment with intrathecal lidocaine given before and after formalin on both nociception and Fos expression in the spinal cord dorsal horn. Lidocaine 56-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 9426840-0 1998 Expression of c-Fos protein immunoreactivity in rat brain during ethanol withdrawal is prevented by nifedipine. Ethanol 65-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 9426840-0 1998 Expression of c-Fos protein immunoreactivity in rat brain during ethanol withdrawal is prevented by nifedipine. Nifedipine 100-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 9426840-2 1998 Immunostaining for c-Fos appeared 2 h after ethanol withdrawal, the number of cells increased significantly at 8 h, but c-Fos had returned to basal level after 24 h. Immunoreactive cells were distributed throughout the brain but were concentrated in cerebral cortex, striatum, and hippocampus. Ethanol 44-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 9426840-3 1998 Intraperitoneal injection of the calcium channel antagonist nifedipine (3 x 100 mg/kg) prior to, and during ethanol withdrawal totally prevented c-Fos protein-like expression. Nifedipine 60-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-150 9426840-4 1998 These results suggest that the superinduction of c-Fos protein in the brain of rats undergoing ethanol withdrawal is induced by calcium influx into neuronal cells, and may be related to previously reported increases in L-type voltage-operated calcium channels in the brain associated with ethanol dependence. Ethanol 95-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 9426840-4 1998 These results suggest that the superinduction of c-Fos protein in the brain of rats undergoing ethanol withdrawal is induced by calcium influx into neuronal cells, and may be related to previously reported increases in L-type voltage-operated calcium channels in the brain associated with ethanol dependence. Calcium 128-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 9426840-4 1998 These results suggest that the superinduction of c-Fos protein in the brain of rats undergoing ethanol withdrawal is induced by calcium influx into neuronal cells, and may be related to previously reported increases in L-type voltage-operated calcium channels in the brain associated with ethanol dependence. Ethanol 289-296 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 9447868-3 1998 METHODS: Formalin-induced nociception and Fos expression in the spinal cord, in response to a 50-microl injection of 2.5% formalin into the hind paw, were assessed in rats given an intrathecal injection of 50 microl 2% lidocaine by lumbar puncture between the L5 and L6 vertebrae, either 3 min before (pretreatment) or 5 min after (post-treatment) formalin injection. Formaldehyde 122-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 9447868-7 1998 On the other hand, the finding that the intrathecal injection of lidocaine after formalin treatment reduced Fos expression but not nociceptive responses indicates an uncoupling of the behavioral and Fos protein responses to formalin and suggests that changes in Fos expression may not be a good predictor of the ability of agents to produce preemptive analgesia. Lidocaine 65-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 9447868-7 1998 On the other hand, the finding that the intrathecal injection of lidocaine after formalin treatment reduced Fos expression but not nociceptive responses indicates an uncoupling of the behavioral and Fos protein responses to formalin and suggests that changes in Fos expression may not be a good predictor of the ability of agents to produce preemptive analgesia. Lidocaine 65-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-202 9447868-7 1998 On the other hand, the finding that the intrathecal injection of lidocaine after formalin treatment reduced Fos expression but not nociceptive responses indicates an uncoupling of the behavioral and Fos protein responses to formalin and suggests that changes in Fos expression may not be a good predictor of the ability of agents to produce preemptive analgesia. Lidocaine 65-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-202 9447868-7 1998 On the other hand, the finding that the intrathecal injection of lidocaine after formalin treatment reduced Fos expression but not nociceptive responses indicates an uncoupling of the behavioral and Fos protein responses to formalin and suggests that changes in Fos expression may not be a good predictor of the ability of agents to produce preemptive analgesia. Formaldehyde 81-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 9473635-5 1998 MK801 pretreatment produced a very strong inhibition of Fos, Jun-D and Krox-20 increases in dentate neurons but had a much smaller effect on Jun-B and c-Jun expression. Dizocilpine Maleate 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 9473567-0 1998 2,5-Anhydro-D-mannitol induces Fos-like immunoreactivity in hindbrain and forebrain: relationship to eating behavior. 2,5-anhydromannitol 0-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 9473567-2 1998 Previous studies have shown that administration of 2,5-AM in doses that elicit eating induces Fos-like immunoreactivity (Fos-li) primarily in hindbrain structures, including the nucleus of the solitary tract (NTS), area postrema (AP), and lateral parabrachial nucleus (PBN). 2,5-anhydromannitol 51-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 9473567-2 1998 Previous studies have shown that administration of 2,5-AM in doses that elicit eating induces Fos-like immunoreactivity (Fos-li) primarily in hindbrain structures, including the nucleus of the solitary tract (NTS), area postrema (AP), and lateral parabrachial nucleus (PBN). 2,5-anhydromannitol 51-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-124 9473567-5 1998 Doses of 2,5-AM that reliably stimulated food intake induced Fos-li in both the hindbrain and forebrain, including in the NTS, AP, lateral PBN, central lateral nucleus of the amygdala, dorsal lateral bed nucleus of the stria terminalis (BNSTdl), anterior paraventricular nucleus of the thalamus, supraoptic nucleus, subfornical organ, and paraventricular hypothalamic nuclei. 2,5-anhydromannitol 9-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 9473572-0 1998 Neuroanatomical patterns of Fos-like immunoreactivity induced by naltrexone in food-restricted and ad libitum fed rats. Naltrexone 65-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 9473572-3 1998 In the present study, c-Fos immunohistochemistry was used to localize cells that are released from opioid-mediated inhibition by naltrexone under conditions of food restriction and ad libitum feeding. Naltrexone 129-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 9510424-0 1998 Opiate disruption of maternal behavior: morphine reduces, and naloxone restores, c-fos activity in the medial preoptic area of lactating rats. Opiate Alkaloids 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 9510424-0 1998 Opiate disruption of maternal behavior: morphine reduces, and naloxone restores, c-fos activity in the medial preoptic area of lactating rats. Naloxone 62-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 9510424-3 1998 In two experiments, the effects of morphine-alone and morphine plus naloxone on the expression of c-fos were examined. Morphine 35-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 9510424-3 1998 In two experiments, the effects of morphine-alone and morphine plus naloxone on the expression of c-fos were examined. Morphine 54-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 9510424-3 1998 In two experiments, the effects of morphine-alone and morphine plus naloxone on the expression of c-fos were examined. Naloxone 68-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 9510424-7 1998 Morphine-treated females had fewer c-fos cells in mPOA compared to saline-treated females, and the presence of pups accounted for a significant increase in c-fos-expressing neurons, whereas in females not exposed to pups, morphine treatment did not significantly reduce baseline c-fos expression (experiment 1). Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 9510424-7 1998 Morphine-treated females had fewer c-fos cells in mPOA compared to saline-treated females, and the presence of pups accounted for a significant increase in c-fos-expressing neurons, whereas in females not exposed to pups, morphine treatment did not significantly reduce baseline c-fos expression (experiment 1). Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 9510424-7 1998 Morphine-treated females had fewer c-fos cells in mPOA compared to saline-treated females, and the presence of pups accounted for a significant increase in c-fos-expressing neurons, whereas in females not exposed to pups, morphine treatment did not significantly reduce baseline c-fos expression (experiment 1). Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 9510424-8 1998 Furthermore, naloxone mitigated the effect as morphine + naloxone-treated females expressed more c-fos cells compared to morphine + saline females (experiment 2). Naloxone 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 9510424-8 1998 Furthermore, naloxone mitigated the effect as morphine + naloxone-treated females expressed more c-fos cells compared to morphine + saline females (experiment 2). Morphine 46-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 9510424-8 1998 Furthermore, naloxone mitigated the effect as morphine + naloxone-treated females expressed more c-fos cells compared to morphine + saline females (experiment 2). Naloxone 57-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 9507053-10 1998 In dMEA and CTF, nuclear colocalization of AR-ir and mating-induced Fos-ir was present in a proportion of FG-containing neurons. 2-(dimethylamino)ethanol 3-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 9507053-10 1998 In dMEA and CTF, nuclear colocalization of AR-ir and mating-induced Fos-ir was present in a proportion of FG-containing neurons. CHEMBL408967 12-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 9473635-10 1998 Conversely, the role in kindling of those genes whose expression was significantly attenuated by MK801 (Fos, Jun-D, Krox-20, trkB and BDNF) requires further examination. Dizocilpine Maleate 97-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 9421421-1 1998 c-Fos/c-Jun dimers (activating protein-1 transcription factor) are involved in the modulatory actions of angiotensin II (Ang II) on brain norepinephrine neurons, effects mediated via Ang II type 1 (AT1) receptors. Norepinephrine 138-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9454625-4 1998 After 5 x ECS and 10 x ECS, c-Fos was reexpressed in the CA4 area, but was completely absent in the other hippocampal areas and cortex. 2-Hydroxyestradiol 10-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 9502114-0 1998 Involvement of Ca2+ signalling in the vasoactive intestinal peptide and 8-Br-cAMP induction of c-fos mRNA expression. 8-Bromo Cyclic Adenosine Monophosphate 72-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 9502114-2 1998 VIP stimulated c-fos mRNA expression in a clonal GH3 pituitary tumour cell line, GH3Ca, whereas 8-Br-cAMP only moderately induced c-fos expression. 8-Bromo Cyclic Adenosine Monophosphate 96-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 9502114-3 1998 The VIP-induced c-fos expression was inhibited in the presence of EGTA, or the L-type Ca2+ channel blockers verapamil and nifedipine. Egtazic Acid 66-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 9502114-3 1998 The VIP-induced c-fos expression was inhibited in the presence of EGTA, or the L-type Ca2+ channel blockers verapamil and nifedipine. Verapamil 108-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 9502114-3 1998 The VIP-induced c-fos expression was inhibited in the presence of EGTA, or the L-type Ca2+ channel blockers verapamil and nifedipine. Nifedipine 122-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 9502114-9 1998 Interestingly, pretreatment with thapsigargin greatly potentiated the 8-Br-cAMP-stimulated c-fos expression. Thapsigargin 33-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 9502114-9 1998 Interestingly, pretreatment with thapsigargin greatly potentiated the 8-Br-cAMP-stimulated c-fos expression. 8-Bromo Cyclic Adenosine Monophosphate 70-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 9495582-0 1998 Dose-related anti-inflammatory/analgesic effects of lornoxicam: a spinal c-Fos protein study in the rat. lornoxicam 52-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 9495582-3 1998 RESULTS: Lornoxicam dose-relatedly reduced both the carrageenan evoked oedema (r=0.63 and r=0.53 for paw and ankle diameter respectively; p<0.001 for both) and total number of spinal c-Fos-LI neurons (r=0.79; p<0.001), with the strongest effect corresponding to a 75 +/- 2% reduction of the number of c-Fos-LI neurons (p<0.001) for the highest dose (9 mg/kg), and a 45 +/- 3% reduction (p<0.001) for the low dose of 0.3 mg/kg. lornoxicam 9-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 186-191 9495582-3 1998 RESULTS: Lornoxicam dose-relatedly reduced both the carrageenan evoked oedema (r=0.63 and r=0.53 for paw and ankle diameter respectively; p<0.001 for both) and total number of spinal c-Fos-LI neurons (r=0.79; p<0.001), with the strongest effect corresponding to a 75 +/- 2% reduction of the number of c-Fos-LI neurons (p<0.001) for the highest dose (9 mg/kg), and a 45 +/- 3% reduction (p<0.001) for the low dose of 0.3 mg/kg. lornoxicam 9-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 307-312 9495582-5 1998 CONCLUSIONS: Our results demonstrate that lornoxicam reduces in parallel both the carrageenan-evoked oedema and spinal c-Fos expression, with clear evidence for a potent effect of low doses of lornoxicam. lornoxicam 42-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 9495582-5 1998 CONCLUSIONS: Our results demonstrate that lornoxicam reduces in parallel both the carrageenan-evoked oedema and spinal c-Fos expression, with clear evidence for a potent effect of low doses of lornoxicam. lornoxicam 193-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 9453311-8 1998 Lyso-PC (25 micromol/L) also increased the mRNA expression of c-fos and c-jun genes. We 201 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 9454625-4 1998 After 5 x ECS and 10 x ECS, c-Fos was reexpressed in the CA4 area, but was completely absent in the other hippocampal areas and cortex. 2-Hydroxyestradiol 23-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 9440082-11 1998 BAPTA inhibited c-fos expression observed in response to these agents. 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 9495582-6 1998 Correlated reductions in c-Fos expression and paw oedema suggest a predominantly peripheral site of action of lornoxicam. lornoxicam 110-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 9440082-12 1998 Phorbol ester induction of c-fos mRNA in the absence of raised cytosolic or nuclear calcium was also suppressed by BAPTA. Phorbol Esters 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 9440082-12 1998 Phorbol ester induction of c-fos mRNA in the absence of raised cytosolic or nuclear calcium was also suppressed by BAPTA. 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid 115-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 9869327-0 1998 C-fos expression in the rat nucleus basalis upon excitotoxic lesion with quisqualic acid: a study in adult and aged animals. Quisqualic Acid 73-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9435201-8 1998 pretreatment with an equieffective dose of the mu opioid receptor agonist [D-Ala2,NMePhe4,Gly-ol5]enkephalin (DAMGO) not only significantly decreased the number of flinches in phase 1 and phase 2, but also nearly completely prevented the expression of Fos-LI in all regions of the spinal cord. gly-ol5]enkephalin 90-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 252-255 9435201-11 1998 administered DAMGO suggests that a direct spinal action contributes to the inhibition of noxious stimulus-evoked Fos-LI in the spinal cord produced by systemically administered mu opioid receptor agonists such as morphine. Enkephalin, Ala(2)-MePhe(4)-Gly(5)- 13-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-116 9435201-11 1998 administered DAMGO suggests that a direct spinal action contributes to the inhibition of noxious stimulus-evoked Fos-LI in the spinal cord produced by systemically administered mu opioid receptor agonists such as morphine. Morphine 213-221 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-116 9519796-4 1998 The high expression of c-fos gene at 13:00 could be associated to the increase in estradiol seric levels observed both at 11:00 h and at 13:00 h. Our results correlate with the increase in the number of FOS immunoreactive cells in some forebrain areas in proestrus afternoon related to gonadotropin releasing hormone secretion. seric 92-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 9651118-0 1998 Involvement of dopamine D1 and D2 receptors in Fos immunoreactivity induced by stepholidine in both intact and denervated striatum of lesioned rats. stepholidine 79-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 9651118-6 1998 The Fos-positive cells were mainly found in striatal neurons retrogradely labeled with horseradish peroxidase (HRP) from GP but not from SN, and could be abolished by the pretreatment of a D2 agonist LY171555 (2 mg/kg, i.p. 4,4a,5,6,7,8,8a,9-octahydro-5-propyl-1H-pyrzolo(3,4-g)quinoline 200-208 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-7 9651118-10 1998 Similar to that of normal rats, the Fos expression in intact side possessed the rostral-caudal gradient and could be abolished by the pretreatment of LY171555. 4,4a,5,6,7,8,8a,9-octahydro-5-propyl-1H-pyrzolo(3,4-g)quinoline 150-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-39 9483519-10 1998 These data suggest that trigeminal stimuli received by lactating rats during the performance of pronurturant maternal behaviour promote cellular activity resulting in neuronal expression of c-fos in many forebrain sites including the medial preoptic nucleus, several sites connected with it that are part of the mesotelencephalic dopamine system, and in the somatosensory cortex. Dopamine 330-338 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 190-195 9681931-0 1998 Regional differences in the ability of caffeine to affect haloperidol-induced striatal c-fos mRNA expression in the rat. Caffeine 39-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 9681931-0 1998 Regional differences in the ability of caffeine to affect haloperidol-induced striatal c-fos mRNA expression in the rat. Haloperidol 58-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 9681931-1 1998 By using in situ hybridisation we examined the acute effects of caffeine on haloperidol-induced c-fos mRNA in rat striatum. Caffeine 64-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 9681931-1 1998 By using in situ hybridisation we examined the acute effects of caffeine on haloperidol-induced c-fos mRNA in rat striatum. Haloperidol 76-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 9681931-2 1998 A homogeneous induction of striatal c-fos mRNA was found 30 min after injection of haloperidol (1 mg kg(-1)). Haloperidol 83-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 9681931-4 1998 When caffeine was injected together with haloperidol c-fos mRNA was reduced in the medial part of the striatum, but enhanced in the caudal part. Haloperidol 41-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 9681931-5 1998 Similar region-specific effects of caffeine were observed on c-fos mRNA induced by the selective dopamine D2 antagonist raclopride (0.5 mg kg(-1)). Caffeine 35-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 9681931-5 1998 Similar region-specific effects of caffeine were observed on c-fos mRNA induced by the selective dopamine D2 antagonist raclopride (0.5 mg kg(-1)). Dopamine 97-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 9681931-5 1998 Similar region-specific effects of caffeine were observed on c-fos mRNA induced by the selective dopamine D2 antagonist raclopride (0.5 mg kg(-1)). Raclopride 120-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 9681931-6 1998 Both haloperidol and raclopride counteracted caffeine-induced c-fos mRNA expression in somatosensory cortex. Haloperidol 5-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 9681931-6 1998 Both haloperidol and raclopride counteracted caffeine-induced c-fos mRNA expression in somatosensory cortex. Raclopride 21-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 9681931-6 1998 Both haloperidol and raclopride counteracted caffeine-induced c-fos mRNA expression in somatosensory cortex. Caffeine 45-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 9681931-8 1998 The present data show that caffeine modulates c-fos mRNA induced by dopamine D2 receptor antagonism differentially in sensorimotor and limbic-related areas of striatum. Caffeine 27-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 9681934-2 1998 Our study was designed to investigate the effect of lithium on the 5-HT2A or 5-HT2C (5-HT2A/2C) receptor subtypes, by comparing the consequences of chronic pre-treatment of rats with lithium on 5-HT2A/2C receptor-mediated behavioural responses, Fos expression, and the density of these receptors in the brain. Lithium 183-190 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 245-248 9681934-3 1998 In addition, the time-course and persistence of the effect of chronic lithium on 5-HT2A/2C receptor-mediated Fos expression was examined. Lithium 70-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-112 9681934-4 1998 Furthermore, the acute action of lithium on Fos expression was also examined. Lithium 33-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 9681934-5 1998 In an investigation of the dose response of Fos to the 5-HT2A/2C agonist 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI), rats received saline or 1, 2, 4, 8, 12, 16, 24 or 32 mg/kg DOI, then were sacrificed 3 h later for immunocytochemical localisation of Fos. 4-iodo-2,5-dimethoxyphenylisopropylamine 73-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 9681934-15 1998 Treatment of rats with chronic lithium significantly enhanced DOI-induced locomotor activity and Fos-like immunoreactivity throughout the cerebral cortex. Lithium 31-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 9681934-16 1998 This elevation in Fos-like immunoreactivity was completely abolished by prior treatment with ritanserin. Ritanserin 93-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 9681934-18 1998 The results of the time-course experiment demonstrated that the enhancing effect of lithium on 5-HT2A/2C receptor-mediated Fos expression was short-lived such that Fos-like immunoreactivity returned to untreated levels within 48 h. In the acute lithium experiment, administration of lithium to rats 12 or 24 h before DOI resulted in a similar elevation of Fos-like immunoreactivity to that seen in chronically treated animals. Lithium 84-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-126 9681934-18 1998 The results of the time-course experiment demonstrated that the enhancing effect of lithium on 5-HT2A/2C receptor-mediated Fos expression was short-lived such that Fos-like immunoreactivity returned to untreated levels within 48 h. In the acute lithium experiment, administration of lithium to rats 12 or 24 h before DOI resulted in a similar elevation of Fos-like immunoreactivity to that seen in chronically treated animals. Lithium 84-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-167 9681934-18 1998 The results of the time-course experiment demonstrated that the enhancing effect of lithium on 5-HT2A/2C receptor-mediated Fos expression was short-lived such that Fos-like immunoreactivity returned to untreated levels within 48 h. In the acute lithium experiment, administration of lithium to rats 12 or 24 h before DOI resulted in a similar elevation of Fos-like immunoreactivity to that seen in chronically treated animals. Lithium 84-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-167 9681934-18 1998 The results of the time-course experiment demonstrated that the enhancing effect of lithium on 5-HT2A/2C receptor-mediated Fos expression was short-lived such that Fos-like immunoreactivity returned to untreated levels within 48 h. In the acute lithium experiment, administration of lithium to rats 12 or 24 h before DOI resulted in a similar elevation of Fos-like immunoreactivity to that seen in chronically treated animals. Lithium 245-252 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-126 9681934-18 1998 The results of the time-course experiment demonstrated that the enhancing effect of lithium on 5-HT2A/2C receptor-mediated Fos expression was short-lived such that Fos-like immunoreactivity returned to untreated levels within 48 h. In the acute lithium experiment, administration of lithium to rats 12 or 24 h before DOI resulted in a similar elevation of Fos-like immunoreactivity to that seen in chronically treated animals. Lithium 245-252 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-126 9583573-3 1998 Capsaicin (100 mg/kg subcutaneous (s.c.), 7 days prior to the experiment) which does not block bladder reflexes but does desensitize C-fiber afferents, reduced (89%) the number of Fos-positive cells in the lumbosacral spinal cord induced by acetic acid-induced irritation of the LUT. Capsaicin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 180-183 9435201-2 1998 administered mu and delta opioid receptor agonists on the flinching behavior and the expression of Fos-like immunoreactivity (Fos-LI) in the spinal cord elicited by s.c. injection of 5% formalin in one hindpaw of the rat. Formaldehyde 186-194 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 9435201-2 1998 administered mu and delta opioid receptor agonists on the flinching behavior and the expression of Fos-like immunoreactivity (Fos-LI) in the spinal cord elicited by s.c. injection of 5% formalin in one hindpaw of the rat. Formaldehyde 186-194 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-129 9435201-5 1998 pretreatment with 60 micrograms of DPDPE produced a small decrease in the numbers of Fos-LI neurons in laminae I, IIi and IIo, as well as laminae V and VI and laminae VII-X, i.t. Enkephalin, D-Penicillamine (2,5)- 35-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 9583573-10 1998 The ganglionic blocking agent, hexamethonium, which blocks autonomic but not afferent pathways to the LUT, decreased c-fos expression by 50%. Hexamethonium 31-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 9459544-9 1997 Double labeling of c-Fos and PRs in progesterone-facilitated rats indicated that nearly all the c-Fos-positive neurons of the pePOA (80 +/- 4.2%) co-expressed PRs; in progesterone-inhibited rats, only 32 +/- 12% of few c-Fos-positive neurons also contained PRs. Progesterone 36-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 9495545-1 1997 We have assessed whether melatonin can induce c-fos expression at various circadian phases, and whether melatonin can inhibit photically induced c-fos expression in the suprachiasmatic nucleus (SCN) in both rats and Syrian hamsters. Melatonin 104-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-150 9495545-2 1997 Subcutaneous administration of melatonin at a dose of 100 microg/kg neither induced expression of Fos, the protein product of the c-fos proto-oncogene, nor inhibited the expression of Fos-like immunoreactivity (Fos-lir) induced by a light pulse in the SCN of rats and hamsters. Melatonin 31-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 184-187 9495545-2 1997 Subcutaneous administration of melatonin at a dose of 100 microg/kg neither induced expression of Fos, the protein product of the c-fos proto-oncogene, nor inhibited the expression of Fos-like immunoreactivity (Fos-lir) induced by a light pulse in the SCN of rats and hamsters. Melatonin 31-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 184-187 9459544-9 1997 Double labeling of c-Fos and PRs in progesterone-facilitated rats indicated that nearly all the c-Fos-positive neurons of the pePOA (80 +/- 4.2%) co-expressed PRs; in progesterone-inhibited rats, only 32 +/- 12% of few c-Fos-positive neurons also contained PRs. Progesterone 36-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 9459544-9 1997 Double labeling of c-Fos and PRs in progesterone-facilitated rats indicated that nearly all the c-Fos-positive neurons of the pePOA (80 +/- 4.2%) co-expressed PRs; in progesterone-inhibited rats, only 32 +/- 12% of few c-Fos-positive neurons also contained PRs. Progesterone 36-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 9547162-0 1997 Developmental changes in distribution patterns of phencyclidine-induced c-Fos in rat forebrain. Phencyclidine 50-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 9466718-1 1997 We have used immunocytochemical detection of c-fos expression (FOS-like immunoreactivity, FLI) to establish the site of action of monosodium glutamate (MSG) in neonatal rats in a model of lesion-induced precocious puberty. Sodium Glutamate 152-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 9466718-11 1997 The receptor which mediates MSG-induced c-fos expression, in the presence of MK-801 or DNQX, needs to be identified. Dizocilpine Maleate 77-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 9466718-11 1997 The receptor which mediates MSG-induced c-fos expression, in the presence of MK-801 or DNQX, needs to be identified. FG 9041 87-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 9547162-1 1997 In the forebrain of 56-day-old rats, histochemical studies revealed that the subcutaneous injection of a psychotomimetic phencyclidine (PCP; 1 and 10 mg/kg) induced a dose-related and dense nuclear c-Fos-like immunoreactivity in the pyriform cortex, layers IV-VI of the neocortex and septum, but a sparse c-Fos immunostaining in the olfactory tubercle and mid-lateral striatum. Phencyclidine 121-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 198-203 9547162-1 1997 In the forebrain of 56-day-old rats, histochemical studies revealed that the subcutaneous injection of a psychotomimetic phencyclidine (PCP; 1 and 10 mg/kg) induced a dose-related and dense nuclear c-Fos-like immunoreactivity in the pyriform cortex, layers IV-VI of the neocortex and septum, but a sparse c-Fos immunostaining in the olfactory tubercle and mid-lateral striatum. Phencyclidine 121-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 305-310 9547162-1 1997 In the forebrain of 56-day-old rats, histochemical studies revealed that the subcutaneous injection of a psychotomimetic phencyclidine (PCP; 1 and 10 mg/kg) induced a dose-related and dense nuclear c-Fos-like immunoreactivity in the pyriform cortex, layers IV-VI of the neocortex and septum, but a sparse c-Fos immunostaining in the olfactory tubercle and mid-lateral striatum. pcp 136-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 198-203 9547162-1 1997 In the forebrain of 56-day-old rats, histochemical studies revealed that the subcutaneous injection of a psychotomimetic phencyclidine (PCP; 1 and 10 mg/kg) induced a dose-related and dense nuclear c-Fos-like immunoreactivity in the pyriform cortex, layers IV-VI of the neocortex and septum, but a sparse c-Fos immunostaining in the olfactory tubercle and mid-lateral striatum. pcp 136-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 305-310 9547162-2 1997 Infant rats at postnatal day 8 expressed much fewer and more confined c-Fos-positive cells in the neocortex than young adult rats following PCP injection. pcp 140-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 9547162-4 1997 These developmental changes in the regional distribution of a neuronal activity marker, c-Fos, suggest that neuronal populations involved in PCP-induced abnormal behavior are influenced by postnatal development, at least, in the neocortex. pcp 141-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 9462893-14 1997 Nifedipine was found to suppress both Ang II-induced corticosterone release and c-fos expression in the following areas: organum vasculosum of the lamina terminalis (OVLT), median preoptic nucleus (MNPO), hypothalamic paraventricular nucleus (PVN) and supraoptic nucleus (SON). Nifedipine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 9461033-7 1997 All four compounds increase uterine VEGF and c-fos mRNA levels indicating that the triphenylethylene class of antiestrogens are predominantly agonists for the induction of these genes in the uterus. triphenylethylene 83-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 9673010-4 1997 To this aim, groups of intact (IN), adrenalectomized (ADX) and sham-operated (sham) rats were killed 7 days after surgery (or no surgery) at times when Fos-IR is known to show either nadir (at light offset) or peak (6 h after light offset) values within MBH POMC neurons. nadir 183-188 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-155 9454666-3 1997 Herein we further evaluated the effects and site-specificity of various somatosensory cues received from pups during 60 min on Fos-ir in the PAG of day 7 postpartum rats after a 48-h dam-litter separation. phenylacetylglycine 141-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-130 9450688-0 1997 Principal involvement of cyclooxygenase-1-derived prostaglandins in the c-fos expression of the rat hind brain following visceral stimulation with acetic acid. Prostaglandins 50-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 9450688-0 1997 Principal involvement of cyclooxygenase-1-derived prostaglandins in the c-fos expression of the rat hind brain following visceral stimulation with acetic acid. Acetic Acid 147-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 9454666-4 1997 Dams interacting with suckling versus nonsuckling pups showed relatively high numbers of Fos-ir cells in the intercollicular cPAG site identified earlier, but not in three other rostrocaudal planes of the PAG. cpag 125-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 9454666-6 1997 Perioral anesthesia of dams prior to reunion with the litter prevented retrieval and licking of pups but not pup-initiated nursing behavior, the duration of which was positively correlated with Fos-ir levels within the intercollicular cPAG. cpag 235-239 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 194-197 9454666-7 1997 Thus, various somatosensory stimuli from pups activate c-fos in a discrete region of the cPAG but only interactions that include suckling and its behavioral consequences elicit maximal expression, consistent with a role for this midbrain site in the sensorimotor control of kyphotic nursing in rats. cpag 89-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 9418962-2 1997 Activation of this neurotransmitter receptor by the stable acetylcholine analog carbachol (CCh) triggers transducing events, modulating c-fos expression and cellular proliferation. Acetylcholine 59-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 9375664-5 1997 Gel supershift assays using transcription factor-specific antibodies revealed that p-CREB, Jun D, and a Fos family protein(s) are components of the AP-1 binding complex in untreated and lithium-treated CGC. Lithium 186-193 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 9375664-7 1997 Similar to the results obtained in CGC, p-CREB, Jun D, and Fos family proteins are present in the AP-1 binding sites in the frontal cortex and hippocampus of untreated and lithium-treated rats. Lithium 172-179 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 9418962-2 1997 Activation of this neurotransmitter receptor by the stable acetylcholine analog carbachol (CCh) triggers transducing events, modulating c-fos expression and cellular proliferation. Carbachol 80-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 9418962-2 1997 Activation of this neurotransmitter receptor by the stable acetylcholine analog carbachol (CCh) triggers transducing events, modulating c-fos expression and cellular proliferation. Carbachol 91-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 9469524-0 1997 Formalin-induced c-fos expression in the spinal cord of fetal rats. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 9330358-0 1997 Sex-dependent effects of formalin and restraint on c-Fos expression in the septum and hippocampus of the rat. Formaldehyde 25-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 9330358-5 1997 In both male and female rats, unilateral injection of formalin induced bilateral c-Fos expression in the hippocampus, but the number of labeled neurons was two-fold higher in females than in males. Formaldehyde 54-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 9330360-7 1997 c-Fos immunohistochemistry was used routinely to provide regional estimates of the suppressive effects of muscimol on neuronal activity. Muscimol 106-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9473144-0 1997 The preferential dopamine D3 receptor agonist cis-8-OH-PBZI induces limbic Fos expression in rat brain. 8-hydroxy-3-(n-propyl)-1,2,3a,4,5,9b-hexahydro-1H-benz(e)indole 46-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 9473144-4 1997 In vivo, cis-8-OH-PBZI potently induced Fos protein immunoreactivity in the rat medial prefrontal cortex and shell region of the nucleus accumbens, but only marginally in the motor dorsolateral striatum, indicating a selective limbic site of action. 8-hydroxy-3-(n-propyl)-1,2,3a,4,5,9b-hexahydro-1H-benz(e)indole 9-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 9450688-3 1997 Pharmacological experiments using mofezolac, a preferential inhibitor against COX-1, and NS-398, a selective inhibitor against COX-2, confirmed the involvement of COX-1 derived PGs in the induction of c-fos expression in the hind brain following the noxious stimulation. mofezolac 34-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 201-206 9450688-3 1997 Pharmacological experiments using mofezolac, a preferential inhibitor against COX-1, and NS-398, a selective inhibitor against COX-2, confirmed the involvement of COX-1 derived PGs in the induction of c-fos expression in the hind brain following the noxious stimulation. N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide 89-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 201-206 9450688-3 1997 Pharmacological experiments using mofezolac, a preferential inhibitor against COX-1, and NS-398, a selective inhibitor against COX-2, confirmed the involvement of COX-1 derived PGs in the induction of c-fos expression in the hind brain following the noxious stimulation. Phosphatidylglycerols 177-180 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 201-206 9542774-0 1997 c-fos expression in the rat hypothalamic paraventricular nucleus induced by LiCl: descending projections to the dorsal vagal motor nucleus. Lithium Chloride 76-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9396461-9 1997 Northern blot analysis indicated that bcl-2 and c-fos but not p53 and c-myc may participate in mediating H2O2-Fe(II)-induced VSMC apoptosis. ammonium ferrous sulfate 110-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 9469524-1 1997 The maturational status of Adelta and C-fibers in the fetal rat spinal cord was examined using formalin-induced c-fos expression as a marker for neuronal activities. Formaldehyde 95-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 9469524-5 1997 The anatomical data paralleled that of behavior; FD 19 animals expressed a small number of Fos labeled nuclei following the formalin injection that was not statistically different from control animals. Formaldehyde 124-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-94 9469524-6 1997 The formalin-induced increase in Fos staining was first observed at FD 20 with a large increase in the number of Fos labeled cell occurring between FD 20 and 21. Formaldehyde 4-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 9469524-6 1997 The formalin-induced increase in Fos staining was first observed at FD 20 with a large increase in the number of Fos labeled cell occurring between FD 20 and 21. Formaldehyde 4-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-116 9396461-9 1997 Northern blot analysis indicated that bcl-2 and c-fos but not p53 and c-myc may participate in mediating H2O2-Fe(II)-induced VSMC apoptosis. Hydrogen Peroxide 105-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 9469524-9 1997 Formalin treated fetuses showed constitutive level of staining in addition to the increase in the c-fos expression caused by formalin. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 9469524-9 1997 Formalin treated fetuses showed constitutive level of staining in addition to the increase in the c-fos expression caused by formalin. Formaldehyde 125-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 9374754-0 1997 Enhancement of spontaneous baroreflex by antisense c-fos oligonucleotide treatment in the NTS of the rat. Oligonucleotides 57-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 9374754-2 1997 In adult male Sprague-Dawley rats anesthetized and maintained with pentobarbital sodium, microinjection bilaterally into the caudal NTS of a 15-mer antisense oligonucleotide that targets against the initiation codon of c-fos mRNA (5"-129 to 143-3") significantly enhanced the spontaneous BRR response, as determined by transfer function analysis of SAP and heart rate signals. Oligonucleotides 158-173 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 219-224 9348187-4 1997 Despite the minimal HPA axis response in nondeprived rats during the SHRP (postnatal day 12), the mild stress of a saline injection significantly increased messenger RNA levels of two immediate-early genes (IEGs), c-fos and NGFI-B, in the hypothalamic paraventricular nucleus (PVN) and in the cerebral cortex. Sodium Chloride 115-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 214-219 9452191-4 1997 These findings suggest that SCH 23390-induced behavioral supersensitivity and the increased striatal c-fos levels are concomitant but unrelated phenomena. SCH 23390 28-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 9452197-0 1997 p-Chlorophenylalanine and fluoxetine inhibit D-fenfluramine-induced Fos expression in the paraventricular nucleus, cingulate cortex and frontal cortex but not in other forebrain and brainstem regions. Fenclonine 0-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 9452197-0 1997 p-Chlorophenylalanine and fluoxetine inhibit D-fenfluramine-induced Fos expression in the paraventricular nucleus, cingulate cortex and frontal cortex but not in other forebrain and brainstem regions. Fluoxetine 26-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 9452197-0 1997 p-Chlorophenylalanine and fluoxetine inhibit D-fenfluramine-induced Fos expression in the paraventricular nucleus, cingulate cortex and frontal cortex but not in other forebrain and brainstem regions. Dexfenfluramine 45-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 9452197-2 1997 Brain sites activated by D-fenfluramine have been mapped via the expression of the immediate early gene Fos. Dexfenfluramine 25-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 9464938-2 1997 Osmotic stimulation of rats produced by graded infusions of saline at different tonicities was found to lead to the induction of c-fos, nur77 and egr1 mRNAs in magnocellular neurons, as well as in putative afferent neurons, including those in structures of the forebrain (subfornical organ, median preoptic nucleus and organum vasculosum of the lamina terminalis). Sodium Chloride 60-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 9334431-0 1997 Systemic morphine-induced Fos protein in the rat striatum and nucleus accumbens is regulated by mu opioid receptors in the substantia nigra and ventral tegmental area. Morphine 9-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 9334431-1 1997 To characterize how systemic morphine induces Fos protein in dorsomedial striatum and nucleus accumbens (NAc), we examined the role of receptors in striatum, substantia nigra (SN), and ventral tegmental area (VTA). Morphine 29-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 9334431-2 1997 Morphine injected into medial SN or into VTA of awake rats induced Fos in neurons in ipsilateral dorsomedial striatum and NAc. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 9334431-3 1997 Morphine injected into lateral SN induced Fos in dorsolateral striatum and globus pallidus. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 9334431-6 1997 Fos induction in dorsomedial striatum produced by systemic administration of morphine was blocked by (1) SN and VTA injections of the mu1 opioid antagonist naloxonazine and (2) striatal injections of either MK 801, an NMDA glutamate receptor antagonist, or SCH 23390, a D1 dopamine receptor antagonist. Morphine 77-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9334431-6 1997 Fos induction in dorsomedial striatum produced by systemic administration of morphine was blocked by (1) SN and VTA injections of the mu1 opioid antagonist naloxonazine and (2) striatal injections of either MK 801, an NMDA glutamate receptor antagonist, or SCH 23390, a D1 dopamine receptor antagonist. naloxonazine 156-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9334431-6 1997 Fos induction in dorsomedial striatum produced by systemic administration of morphine was blocked by (1) SN and VTA injections of the mu1 opioid antagonist naloxonazine and (2) striatal injections of either MK 801, an NMDA glutamate receptor antagonist, or SCH 23390, a D1 dopamine receptor antagonist. Dizocilpine Maleate 207-213 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9334431-6 1997 Fos induction in dorsomedial striatum produced by systemic administration of morphine was blocked by (1) SN and VTA injections of the mu1 opioid antagonist naloxonazine and (2) striatal injections of either MK 801, an NMDA glutamate receptor antagonist, or SCH 23390, a D1 dopamine receptor antagonist. SCH 23390 257-266 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9334431-7 1997 Fos induction in dorsomedial striatum and NAc after systemic administration of morphine seems to be mediated by dopamine neurons in medial SN and VTA that project to medial striatum and NAc, respectively. Morphine 79-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9334431-7 1997 Fos induction in dorsomedial striatum and NAc after systemic administration of morphine seems to be mediated by dopamine neurons in medial SN and VTA that project to medial striatum and NAc, respectively. Dopamine 112-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9334431-10 1997 This activates D1 dopamine receptors and coupled with the coactivation of NMDA receptors possibly from cortical glutamate input induces Fos in striatal and NAc neurons. Glutamic Acid 112-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-139 9334431-11 1997 The modulation of target gene expression by Fos could influence addictive behavioral responses to opiates. Opiate Alkaloids 98-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 9428617-11 1997 Combination of norepinephrine with pre- and afterload elevation induced the c-fos mRNA signal to appear earlier, to be more pronounced, and to persist for a longer period of time. Norepinephrine 15-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 9300429-6 1997 Carbachol activation of parafascicular neurons also induced the synthesis of c-Fos-like immunoreactive proteins in the pallidal neurons. Carbachol 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 9439680-12 1997 In experiment 2, the effect of 24R,25(OH)2vitamin D3 on the alterations in c-fos, c-myc and c-jun oncogene expression in response to DMH administration was examined by northern blot analysis. 25(oh)2vitamin 35-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 9439680-12 1997 In experiment 2, the effect of 24R,25(OH)2vitamin D3 on the alterations in c-fos, c-myc and c-jun oncogene expression in response to DMH administration was examined by northern blot analysis. 1,2-Dimethylhydrazine 133-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 9436645-5 1997 We further show that prior exposure to IMO stress for 6 days, or implantation of corticosterone pellets suppresses the induction of c-fos, fos B, jun B and NGFI-B, but not that of NGFI-A in the rat PVH. Corticosterone 81-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 9329057-2 1997 administration of glucagon-like peptide-1 (7-36) amide (GLP-1) dose dependently suppresses food intake in rats, and induces activation of c-fos within rat paraventricular hypothalamus (PVN). Amides 49-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 9437800-6 1997 Crocidolite asbestos fibers have been reported to induce sustained expression of the c-fos and c-jun protooncogenes in rat pleural mesothelial cells in vitro (Heintz et al, Proc. Asbestos, Crocidolite 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 9341190-6 1997 Nuclear extracts from PC12 cells display a cAMP-induced complex binding to the DB1 element, and antisera to transcription factors CREB, CREM, Fos, and Jun indicate that these proteins, or closely related family members, interact with DB1. Cyclic AMP 43-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-145 9404938-0 1997 Modulation of c-fos expression in the rat striatum by diazepam. Diazepam 54-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 9404938-2 1997 Since cAMP influences c-fos activity, we examined the effects of diazepam on expression of this immediate early gene, as indicated by Western blot analysis. Cyclic AMP 6-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 9404938-3 1997 Intraperitoneal administration of diazepam increased Fos protein levels in the striatum, but not in the hippocampus. Diazepam 34-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 9404938-6 1997 The possible mechanisms underlying the modulatory effects of diazepam on c-fos expression in the brain are discussed. Diazepam 61-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 9336231-10 1997 In the hypothalamic PVN, inhibition of both c-fos and NGFI-B transcripts by indomethacin was also associated to an abolished influence of the endotoxin on the transcription of neuroendocrine CRF; induction of CRF primary transcript by the middle dose of LPS was selective to the PVN and was completely blocked by pretreatment with indomethacin. Indomethacin 76-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 9336231-10 1997 In the hypothalamic PVN, inhibition of both c-fos and NGFI-B transcripts by indomethacin was also associated to an abolished influence of the endotoxin on the transcription of neuroendocrine CRF; induction of CRF primary transcript by the middle dose of LPS was selective to the PVN and was completely blocked by pretreatment with indomethacin. Indomethacin 331-343 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 9336231-11 1997 Moreover, a large number of tyrosine hydroxylase (TH)-immunoreactive neurons of the VLM (A1/C1) and the NTS (A2/C2) were positive for c-fos mRNA in immune-challenged rats, an effect that was largely prevented by indomethacin in the VLM but not in the NTS. Indomethacin 212-224 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-139 9430419-9 1997 Pretreatment with the muscarinic receptor antagonist methylatropine (i.c.v., 0.5 microg) prevented the pressor response to neostigmine and evoked a reduced Fos-like immunoreactivity compared to animals given neostigmine without methylatropine. methylatropine 53-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-159 9401747-0 1997 Compartmentally specific effects of quinpirole on the striatal Fos expression induced by stimulation of D1-dopamine receptors in intact rats. Quinpirole 36-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 9401747-1 1997 Injections of the full D1-agonist A-77636 (1.45 mg/kg) were found to induce clear Fos-like immunoreactivity (FLI) in the striatum of neurologically intact rats. A 77636 34-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 9406920-0 1997 Effects of pentylenetetrazol-induced status epilepticus on c-Fos and HSP72 immunoreactivity in the immature rat brain. Pentylenetetrazole 11-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 9315922-0 1997 Dopamine-adenosine interactions in the striatum and the globus pallidus: inhibition of striatopallidal neurons through either D2 or A2A receptors enhances D1 receptor-mediated effects on c-fos expression. Dopamine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 187-192 9315922-0 1997 Dopamine-adenosine interactions in the striatum and the globus pallidus: inhibition of striatopallidal neurons through either D2 or A2A receptors enhances D1 receptor-mediated effects on c-fos expression. Adenosine 9-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 187-192 9315922-5 1997 Conversely, the D2 agonist, quinelorane (2 mg/kg), decreased c-fos mRNA in these populations but increased it in globus pallidus. quinelorane 28-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 9315922-6 1997 The adenosine A2A receptor antagonist, SCH 58261 (5 mg/kg), also decreased c-fos mRNA in D2 receptor-containing neurons in striatum but did not affect pallidal c-fos mRNA. 5-amino-7-(2-phenylethyl)-2-(2-furyl)pyrazolo(4,3-e)-1,2,4-triazolo(1,5-c)pyrimidine 39-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 9331175-2 1997 One hour after a formalin injection into the lip, many FOS-immunoreactive cells were seen in the parabrachial nucleus, preferentially on the side ipsilateral to the injection site. Formaldehyde 17-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 9331175-4 1997 In contrast, a formalin injection into the hindpaw resulted in dense FOS-labeling in the superior, dorsal, and central lateral subnuclei, with sparse to moderate labeling in the Kolliker-Fuse nucleus, and sparse labeling in the external lateral and external medial subnuclei, as described previously (Hermanson and Blomqvist, J. Comp. Formaldehyde 15-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 9372217-3 1997 To test this, the effects of the D1 agonist SKF 38393 and D2/3 agonist quinelorane were examined on expression of the immediate early gene products Fos and its related antigens (FRA) in SS-immunoreactive (IR) neurons in the PeVN. quinelorane 71-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-151 9372230-0 1997 Peripheral administration of cholecystokinin activates c-fos expression in the locus coeruleus/subcoeruleus nucleus, dorsal vagal complex and paraventricular nucleus via capsaicin-sensitive vagal afferents and CCK-A receptors in the rat. Capsaicin 170-179 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 9372230-9 1997 Perivagal capsaicin pretreatment diminished the CCK-induced increase in the number of c-Fos-LI-positive cells in the LC/SC by 65%. Capsaicin 10-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 9372230-12 1997 Capsaicin diminished the CCK-induced increase in c-Fos-LI-positive cells in the PVN by 64%, in the NTS by 60%, but in the AP only by 25%. Capsaicin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 9372230-13 1997 Immunostaining against the nuclear antigen c-Fos and the cytoplasmatic antigen tyrosine hydroxylase (TH) showed that 40% of all c-Fos-LI-positive cells in the LC/SC were TH-LI positive at 25 micrograms CCK/kg. th-li 170-175 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-133 9387892-2 1997 Direct comparison of the MDD amplification profiles of duplicate, total RNA samples from the caudate putamen (CPu) of either vehicle or cocaine treated Sprague-Dawley rats indicated that the relative induction of a 240 bp (8G247) product, likely to represent c-fos mRNA, closely paralleled changes in c-fos mRNA as measured by Northern blot analysis. Cocaine 136-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 259-264 9387892-2 1997 Direct comparison of the MDD amplification profiles of duplicate, total RNA samples from the caudate putamen (CPu) of either vehicle or cocaine treated Sprague-Dawley rats indicated that the relative induction of a 240 bp (8G247) product, likely to represent c-fos mRNA, closely paralleled changes in c-fos mRNA as measured by Northern blot analysis. Cocaine 136-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 301-306 9357854-0 1997 Differential induction of c-fos, c-jun, and apoptosis in lung epithelial cells exposed to ROS or RNS. ros 90-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 9357854-0 1997 Differential induction of c-fos, c-jun, and apoptosis in lung epithelial cells exposed to ROS or RNS. Radon 97-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 9357854-6 1997 generated by spermine 1,3-propanediamine N-14-[1-(3-aminopropyl)-2-hydroxy-2-nitrosohydrazino]-butyl] or S-nitroso-N-acetylpenicillamine as well as H2O2 cause increased c-fos and c-jun mRNA levels, nuclear proteins, and complexes binding the activator protein-1 recognition sequence in RLE cells. spermine 1,3-propanediamine 13-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-174 9357854-6 1997 generated by spermine 1,3-propanediamine N-14-[1-(3-aminopropyl)-2-hydroxy-2-nitrosohydrazino]-butyl] or S-nitroso-N-acetylpenicillamine as well as H2O2 cause increased c-fos and c-jun mRNA levels, nuclear proteins, and complexes binding the activator protein-1 recognition sequence in RLE cells. n-14-[1-(3-aminopropyl)-2-hydroxy-2-nitrosohydrazino]-butyl 41-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-174 9357854-6 1997 generated by spermine 1,3-propanediamine N-14-[1-(3-aminopropyl)-2-hydroxy-2-nitrosohydrazino]-butyl] or S-nitroso-N-acetylpenicillamine as well as H2O2 cause increased c-fos and c-jun mRNA levels, nuclear proteins, and complexes binding the activator protein-1 recognition sequence in RLE cells. Hydrogen Peroxide 148-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-174 9549051-9 1997 CNA CRH neurons were most responsive and were maximally stimulated by the low dose of nicotine (62% of CRH neurons were cFos+, compared to 10-27% of the CRH population in other regions, including the PVN). Nicotine 86-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-124 9344561-2 1997 In addition, antibodies directed against the FOS protein, which is generated by activation of the immediate early gene c-fos and is temporally associated with ongoing seizure activity, were used to identify transneuronal pathways activated after kainate-induced seizures (KIS). Kainic Acid 246-253 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 9344561-5 1997 Our results demonstrate a specific pattern of FOS-LI induced by kainate injection. Kainic Acid 64-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 9344561-8 1997 The increase in betaAPP-LI glia was far more extensive in adult than in young rats and the anatomical distribution of betaAPP-LI glia was grossly correlated with FOS-LI. betaapp-li 16-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-165 9344561-10 1997 It is likely that the sequence of events following kainate injection is initially triggered by c-fos gene expression, which is rapidly followed by modulation of betaAPP synthesis in parallel to, or preceding, morphological changes of both microglia and astrocytes. Kainic Acid 51-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 9344561-11 1997 The present study, which extensively characterized early changes in c-fos expression and betaAPP-LI in glia following kainate-induced seizures, is a potentially useful animal model for the in vivo study of numerous facets of betaAPP synthesis and the possible role of such processes in Alzheimer"s disease. Kainic Acid 118-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 9386008-0 1997 Fos expression in the rat parabrachial and Kolliker-Fuse nuclei after electrical stimulation of the trigeminal ethmoidal nerve and water stimulation of the nasal mucosa. Water 131-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9386008-3 1997 To induce the expression of Fos protein, two kinds of stimuli and experimental controls were performed in chloralose/urethane-anesthetized animals: (i) electrical stimulation of the trigeminal ethmoidal nerve (EN5) and, as sham controls, dissection of the EN5 without electrical stimulation, (ii) stimulation of the nasal mucosa with water and, as control experiments, no stimulation. Chloralose 106-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 9386008-3 1997 To induce the expression of Fos protein, two kinds of stimuli and experimental controls were performed in chloralose/urethane-anesthetized animals: (i) electrical stimulation of the trigeminal ethmoidal nerve (EN5) and, as sham controls, dissection of the EN5 without electrical stimulation, (ii) stimulation of the nasal mucosa with water and, as control experiments, no stimulation. Urethane 117-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 9386008-3 1997 To induce the expression of Fos protein, two kinds of stimuli and experimental controls were performed in chloralose/urethane-anesthetized animals: (i) electrical stimulation of the trigeminal ethmoidal nerve (EN5) and, as sham controls, dissection of the EN5 without electrical stimulation, (ii) stimulation of the nasal mucosa with water and, as control experiments, no stimulation. Water 334-339 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 9386008-5 1997 Differences could be observed rostrally in the PBL and KF, where significantly higher numbers of Fos-positive neurons were present after EN5 versus water stimulation. Water 148-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 9386008-8 1997 In the water-stimulated rats compared with the anesthesia controls, a significantly higher number of Fos-immunoreactive neurons was always observed at all rostrocaudal levels in the KF and in the midlevel PBL. Water 7-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-104 9386008-10 1997 In contrast, after water stimulation Fos-positive neurons were exclusively found in the Sp5C. Water 19-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 9421173-0 1997 Involvement of a c-fos-dependent mechanism in caffeine-induced expression of the preprotachykinin A and neurotensin/neuromedin N genes in rat striatum. Caffeine 46-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 9421173-6 1997 The antisense blockade of c-fos reduced the effect of caffeine on the expression of mRNAs for preprotachykinin A and neurotensin/neuromedin N in the ventrolateral caudate-putamen. Caffeine 54-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 9421173-8 1997 Thus caffeine induction of striatal preprotachykinin A mRNA and neurotensin/neuromedin N mRNA, but not of preproenkephalin mRNA or prodynorphin mRNA, may at least in part be mediated by a pathway involving Fos protein. Caffeine 5-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 206-209 9394224-0 1997 Effects of intrathecal monoamine antagonists on the nociceptive c-Fos expression in a lesioned rat spinal cord. monoamine 23-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 9394224-2 1997 administration of monoamine antagonists on formalin-induced neuronal c-Fos expression in two sides of the lumbar dorsal horn were observed in rats with unilateral transection of the dorsolateral funiculus at T11-12 level. monoamine 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 9394224-2 1997 administration of monoamine antagonists on formalin-induced neuronal c-Fos expression in two sides of the lumbar dorsal horn were observed in rats with unilateral transection of the dorsolateral funiculus at T11-12 level. Formaldehyde 43-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 9394224-4 1997 normal saline (control) and then an equal volume of formalin was injected into the two hindpaws, the number of Fos-like immunoreactive neurons were 44% lower on the side of lumbar dorsal horn with intact dorsolateral funiculus (57 +/- 3.1 vs. 103 +/- 3.8). Formaldehyde 52-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 9394224-6 1997 phentolamine (a non-selective alpha-adrenoceptor antagonist) caused an increase of Fos-like immunoreactive neurons on the intact side so showing only a reduction rate of 23% to the lesioned side (p < .01); 3) pretreatment with i.t. Phentolamine 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 9394224-7 1997 cyproheptadine (a 5-HT-receptor antagonist) caused a similar reduction rate of 21% (p < .01) of Fos-like immunoreactive neurons on the intact side; and 4) combined i.t. Cyproheptadine 0-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 9394224-8 1997 pretreatment with phentolamine and cyproheptadine caused a reduction of Fos-like immunoreactive neurons of only 4% on the intact side, namely, the differences in the number of Fos-like immunoreactive neurons on two sides of the lumbar spinal cord owing to the unilateral dorsolateral funiculus lesion were nearly abolished by i.t. Phentolamine 18-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 9394224-8 1997 pretreatment with phentolamine and cyproheptadine caused a reduction of Fos-like immunoreactive neurons of only 4% on the intact side, namely, the differences in the number of Fos-like immunoreactive neurons on two sides of the lumbar spinal cord owing to the unilateral dorsolateral funiculus lesion were nearly abolished by i.t. Phentolamine 18-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 176-179 9394224-8 1997 pretreatment with phentolamine and cyproheptadine caused a reduction of Fos-like immunoreactive neurons of only 4% on the intact side, namely, the differences in the number of Fos-like immunoreactive neurons on two sides of the lumbar spinal cord owing to the unilateral dorsolateral funiculus lesion were nearly abolished by i.t. Cyproheptadine 35-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 9394224-8 1997 pretreatment with phentolamine and cyproheptadine caused a reduction of Fos-like immunoreactive neurons of only 4% on the intact side, namely, the differences in the number of Fos-like immunoreactive neurons on two sides of the lumbar spinal cord owing to the unilateral dorsolateral funiculus lesion were nearly abolished by i.t. Cyproheptadine 35-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 176-179 9328940-6 1997 Immunoblot analysis showed that lovastatin decreased membrane-bound p21ras and inhibited FCS-induced c-fos and c-jun protein expression. Lovastatin 32-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 9423929-6 1997 All the pharmacological treatments administered before injecting nitroglycerin selectively influenced Fos expression in the different brain nuclei. Nitroglycerin 65-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 9284512-4 1997 In the present study, the facilitative effect of the 5-HT1A receptor agonist 8-OH-DPAT on ejaculatory behavior was used to analyze the pattern of Fos immunoreactivity ejaculation preceded by minimal sexual activity. 8-Hydroxy-2-(di-n-propylamino)tetralin 77-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-149 9284512-7 1997 Males that ejaculated with the first intromission and were treated with a higher dose of 8-OH-DPAT (0.8 mg/kg) exhibited similar clusters of Fos-positive neurons in all areas except the posterodorsal preoptic nucleus. 8-Hydroxy-2-(di-n-propylamino)tetralin 89-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 9300603-10 1997 These results are consistent with studies of Fos induction in normosensitive animals following dopamine agonists and are discussed in terms of changes in basal ganglia output pathway activity. Dopamine 95-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 9331908-0 1997 c-fos identifies GABA-synthesizing barosensitive neurons in caudal ventrolateral medulla. gamma-Aminobutyric Acid 17-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9331908-2 1997 infusion of phenylephrine, evoked expression of the immediate early gene c-fos in discrete groups of brain stem neurons. Phenylephrine 12-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 9369365-1 1997 The aim of the study was to measure the changes in cerebral energy metabolism and c-fos mRNA expression following challenge with heroin in drug-naive rats and in animals previously sensitized to the drug. Heroin 129-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 9374273-0 1997 Fos-like immunoreactivity induced by intraplantar injection of endotoxin and its reduction by morphine. Morphine 94-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9374273-3 1997 injections of endotoxin (ET, 1.25 microg) can induce FLI in the lumbar spinal cord of rats and to assess the effects of morphine injection on c-fos expression. Morphine 120-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-147 9369310-3 1997 The distributions of Fos-positive neurons were restricted rostro-caudally following formalin injection and tissue injury compared to transection of the infraorbital nerve. Formaldehyde 84-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 9369315-9 1997 Subcutaneous injection of DEX resulted in more widespread immunostaining for Fos, which occurred in lateral, as well as medial, loci in the preoptic area and hypothalamus, whereas Jun-li was restricted to only medial sites. Dexamethasone 26-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-80 9369315-13 1997 In addition, the wide pattern of immunolabeling for Fos in the systematically treated group may reflect both central and peripheral (indirect) steroid effects. Steroids 143-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 9324226-0 1997 Differential effect of sensitizing regimen on induction of Fos-protein and locomotor activity elicited by apomorphine in rats. Apomorphine 106-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 9324226-2 1997 The purpose of this study was to determine whether apomorphine induces Fos-protein in animals sensitized by repeated treatment with apomorphine and whether the magnitude of such induction parallels the magnitude of behavioral response observed after different sensitizing paradigms. Apomorphine 51-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 9324226-3 1997 Apomorphine did induce Fos-protein expression in animals pretreated with apomorphine; however, the rats showing highest levels of induction were not those showing the largest behavioral responses. Apomorphine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 9324226-3 1997 Apomorphine did induce Fos-protein expression in animals pretreated with apomorphine; however, the rats showing highest levels of induction were not those showing the largest behavioral responses. Apomorphine 73-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 9376532-0 1997 Neuroprotective role of c-fos antisense oligonucleotide: in vitro and in vivo studies. Oligonucleotides 40-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 9376532-1 1997 We investigated the dose-response and time-course of c-fos antisense oligodeoxynucleotide (ASO) treatment against excitatory amino acid (EAA)-induced neurotoxicity in rat hippocampal neurons. Oligodeoxyribonucleotides 69-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 9376532-1 1997 We investigated the dose-response and time-course of c-fos antisense oligodeoxynucleotide (ASO) treatment against excitatory amino acid (EAA)-induced neurotoxicity in rat hippocampal neurons. 1,5-anhydroglucitol 91-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 9376532-1 1997 We investigated the dose-response and time-course of c-fos antisense oligodeoxynucleotide (ASO) treatment against excitatory amino acid (EAA)-induced neurotoxicity in rat hippocampal neurons. Excitatory Amino Acids 114-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 9376532-1 1997 We investigated the dose-response and time-course of c-fos antisense oligodeoxynucleotide (ASO) treatment against excitatory amino acid (EAA)-induced neurotoxicity in rat hippocampal neurons. Excitatory Amino Acids 137-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 9376532-4 1997 In vivo, bilateral intrahippocampal injections of c-fos ASO (0.025 nmol/site) was neuroprotective when administered 30 min before or after NMDA treatment. 1,5-anhydroglucitol 56-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 9376532-4 1997 In vivo, bilateral intrahippocampal injections of c-fos ASO (0.025 nmol/site) was neuroprotective when administered 30 min before or after NMDA treatment. N-Methylaspartate 139-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 9276071-3 1997 This report demonstrates that a single dose of desipramine (10 or 25 mg/kg), a classical tricyclic antidepressant drug acting on the adrenergic system, induced c-fos and zif268 expression in rat hippocampus without affecting c-jun. Desipramine 47-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 9291101-2 1997 By transient transfection analysis, we demonstrated that the activation of phospholipase A2 (PLA2) and the subsequent production of arachidonic acid (AA) are essential for Rac-induced c-fos SRE activation, implying a critical role for PLA2 in the Rac-signalling pathway to the nucleus. Arachidonic Acid 132-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 184-189 9285642-8 1997 Alpha1-AR-stimulated c-fos induction, which could be blocked by 5-methylurapidil but not by chloroethylclonidine, was attenuated by Ang II preincubation (100 nmol/L, 24 hours). 5-methylurapidil 64-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 9383212-7 1997 However, the induction of the immediate early gene c-fos and microglial activation in the retrosplenial cortex after administration of dizocilpine was not attenuated by pretreatment with rolipram (5 or 10 mg/kg, 15 min before dizocilpine). Dizocilpine Maleate 135-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 9439947-0 1997 Systemic administration of ephedrine induces Fos protein expression in caudate putamen and subthalamic nucleus of rats. Ephedrine 27-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 9439947-1 1997 Systemic administration of ephedrine, an anti-hypotensive agent with central effects, induced dramatic expression of Fos protein in the caudate putamen (CPu) and subthalamic nucleus (STN) of rats. Ephedrine 27-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 9452197-3 1997 However, it is not clear that serotonin release in the brain mediates the effects of D-fenfluramine on Fos expression. Dexfenfluramine 85-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-106 9452197-4 1997 The present study determined whether D-fenfluramine induces the expression of Fos in the brain through the release of serotonin. Dexfenfluramine 37-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 9452197-4 1997 The present study determined whether D-fenfluramine induces the expression of Fos in the brain through the release of serotonin. Serotonin 118-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 9452197-6 1997 Both drugs inhibited D-fenfluramine-induced Fos expression in the cingulate cortex, frontal cortex, and the parvocellular subdivision of the paraventricular nucleus of the hypothalamus. Dexfenfluramine 21-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 9452197-7 1997 Neither drug reduced D-fenfluramine-induced Fos responses in several other brain areas, including the caudate-putamen, amygdala, and brainstem regions such as the lateral parabrachial nucleus and nucleus of the solitary tract. Dexfenfluramine 21-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 9452197-8 1997 These results indicate regional specificity of mechanisms mediating D-fenfluramine-induced Fos expression. Dexfenfluramine 68-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-94 9452197-9 1997 It is likely that D-fenfluramine-induced Fos expression at various sites in the brain is mediated via a combination of serotonin release and other, as yet unidentified, neurotransmitters. Dexfenfluramine 18-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 9452197-9 1997 It is likely that D-fenfluramine-induced Fos expression at various sites in the brain is mediated via a combination of serotonin release and other, as yet unidentified, neurotransmitters. Serotonin 119-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 9300403-0 1997 Differing influences of dopamine agonists and antagonists on Fos expression in identified populations of globus pallidus neurons. Dopamine 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 9300403-1 1997 Dopamine agonists increase the activity of globus pallidus neurons, as shown electrophysiologically and with Fos expression. Dopamine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-112 9300403-3 1997 Similar responses occur in the striatum, where both dopamine agonists and D2 blockade induce Fos, although in separate neuronal populations (i.e. striatonigral and -pallidal). Dopamine 52-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 9300403-8 1997 Overall, dopamine drug treatments induced more Fos in parvalbumin-negative than -positive cells. Dopamine 9-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 9300403-9 1997 However, unlike dopamine agonists, eticlopride induced significant Fos only in the parvalbumin-negative cells. eticlopride 35-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 9300403-10 1997 Dopamine agonist-induced Fos was found in pallidal neurons projecting to each of the target nuclei examined, in both normal and nigrostriatal-lesioned rats. Dopamine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 9300403-11 1997 Eticlopride-induced Fos occurred only in pallidal neurons projecting to the striatum, providing functional evidence for pallidostriatal cells without axon collaterals to other nuclei. eticlopride 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 9300403-14 1997 The smaller Fos responses in parvalbumin-containing cells may be due largely to the calcium buffering by the parvalbumin itself. Calcium 84-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-15 9300403-15 1997 Also, the pattern of Fos expression in pallidostriatal neurons suggests that dopamine regulates activity in these cells differently than in other projection populations. Dopamine 77-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 9298762-0 1997 Kainic acid-induced excitotoxicity is associated with a complex c-Fos and c-Jun response which does not preclude either cell death or survival. Kainic Acid 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 9252233-0 1997 Differential cellular distribution and dynamics of HSP70, cyclooxygenase-2, and c-Fos in the rat brain after transient focal ischemia or kainic acid. Kainic Acid 137-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 9252233-4 1997 At 6 h after kainic acid, some co-localization of Hsp70 with c-Fos and cyclooxygenase-2 was seen in pyramidal hippocampal neurons and superficial cortical layers, however by 24 h such colocalization became rare within the cortex but was partially maintained in the hippocampus. Kainic Acid 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 9252233-5 1997 Cyclooxygenase-2 was seen in many neurons that were also immunoreactive for c-Fos in superficial cortical layers, dentate gyrus and pyramidal cell layer of the hippocampus from 6 h after kainic acid. Kainic Acid 187-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 9272486-3 1997 With the exception of the nucleus accumbens-shell, where amperozide failed to produce statistically significant increases, the regional distribution of Fos immunoreactivity following amperozide was similar to that induced by atypical, but not by typical, antipsychotic drugs. amperozide 183-193 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-155 9272482-0 1997 Chronic nicotine enhances basal and nicotine-induced Fos immunoreactivity preferentially in the medial prefrontal cortex of the rat. Nicotine 8-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 9272486-4 1997 In addition, after amperozide the number of Fos-positive nuclei was higher in the nucleus accumbens than in the dorsolateral striatum, a characteristic that is common to all known atypical antipsychotic agents. amperozide 19-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 9272482-0 1997 Chronic nicotine enhances basal and nicotine-induced Fos immunoreactivity preferentially in the medial prefrontal cortex of the rat. Nicotine 36-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 9284358-5 1997 Fos-immunoreactivity in the supraoptic nucleus, and also in the median preoptic nucleus, organum vasculosum of the lamina terminalis and subfornical organ, which project to the supraoptic nucleus, increased following morphine withdrawal. Morphine 217-225 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9272486-0 1997 The putative atypical antipsychotic drug amperozide preferentially increases c-fos expression in rat medial prefrontal cortex and lateral septum. amperozide 41-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 9272486-1 1997 The effects of acute, systemic administration of the putative atypical antipsychotic drug amperozide on c-fos expression in the rat forebrain were studied by means of Fos immunohistochemistry. amperozide 90-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 9272486-1 1997 The effects of acute, systemic administration of the putative atypical antipsychotic drug amperozide on c-fos expression in the rat forebrain were studied by means of Fos immunohistochemistry. amperozide 90-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 167-170 9272486-2 1997 Amperozide significantly increased the number of Fos-immunoreactive nuclei in the medial prefrontal cortex and the lateral septum but not in the nucleus accumbens (shell or core), the striatum, or the amygdala. amperozide 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 9284358-6 1997 However, retrograde tracing from the supraoptic nucleus showed that, of the neurons in these regions which project to the supraoptic nucleus, only 0.4-7.1% expressed Fos in response to morphine withdrawal. Morphine 185-193 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-169 9284361-0 1997 The contribution of supraspinal, peripheral and intrinsic spinal circuits to the pattern and magnitude of Fos-like immunoreactivity in the lumbar spinal cord of the rat withdrawing from morphine. Morphine 186-194 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 9284361-1 1997 Withdrawal from morphine evokes increases in Fos-like immunoreactivity in the spinal cord, particularly in the superficial dorsal horn, laminae I/II. Morphine 16-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 9284361-2 1997 To determine the origin of the increased Fos-like immunoreactivity, we selectively targeted central or peripheral opioid receptors with naloxone-methiodide, an antagonist that does not cross the blood-brain barrier, or induced withdrawal after eliminating possible sources of input to the superficial dorsal horn. N-methylnaloxone 136-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 9284361-6 1997 spinal transection, unilateral dorsal rhizotomy (L4-S2), neonatal capsaicin treatment or direct intrathecal opioid antagonist injection, induced expression of the Fos protein. Capsaicin 66-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 9284361-10 1997 However, intrathecal injection of naloxone-methiodide increased Fos-like immunoreactivity in laminae I/II and the ventral horn to a greater extent than did subcutaneous injection of naloxone. N-methylnaloxone 34-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 9284361-10 1997 However, intrathecal injection of naloxone-methiodide increased Fos-like immunoreactivity in laminae I/II and the ventral horn to a greater extent than did subcutaneous injection of naloxone. Naloxone 34-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 9313281-12 1997 Morphine, but not sumatriptan, reduced c-fos expression in both the ipsilateral and contralateral TNC by 71% (P < 0.05 and P = 0.19, respectively), confirming that nociceptors have been activated. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 9313281-14 1997 Instead we observed a positive, linear correlation between the number of KCl injections and the extent of c-fos expression in TNC (correlation coefficient r = 0.709, P < 0.05). Potassium Chloride 73-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 9365026-0 1997 Amphetamine sensitization augments amphetamine-induced Fos expression in the lateral habenula. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 9364486-2 1997 A significant induction of c-fos mRNA, but not of any of the other IEGs, was found 2, 4 and 8 hr after a single injection of 50 mg/kg caffeine. Caffeine 134-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 9364486-4 1997 The ability of caffeine to increase pallidal c-fos mRNA expression was mimicked by the dopamine D2/3 receptor agonist quinpirole (1 or 3 mg/kg), whereas the dopamine D2/3 receptor antagonist raclopride (2 mg/kg) was ineffective. Caffeine 15-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 9364486-4 1997 The ability of caffeine to increase pallidal c-fos mRNA expression was mimicked by the dopamine D2/3 receptor agonist quinpirole (1 or 3 mg/kg), whereas the dopamine D2/3 receptor antagonist raclopride (2 mg/kg) was ineffective. Quinpirole 118-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 9364486-6 1997 The caffeine-induced c-fos mRNA expression was not counteracted by concomitant treatment with raclopride. Caffeine 4-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 9364486-9 1997 The fact that pallidal c-fos mRNA expression decreased upon repeated administration of caffeine may be related to the development of tolerance to locomotion stimulation that occurs following chronic caffeine ingestion. Caffeine 87-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 9364486-9 1997 The fact that pallidal c-fos mRNA expression decreased upon repeated administration of caffeine may be related to the development of tolerance to locomotion stimulation that occurs following chronic caffeine ingestion. Caffeine 199-207 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 9364488-5 1997 Theophylline enhanced the upregulation of c-fos and NFGI-A during reperfusion but did not prevent the decrease in adenosine A1 receptor mRNA. Theophylline 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 9380276-18 1997 Intracerebroventricular sandostatin injection attenuated the GHRP-6-induced Fos response, from 53 +/- 6 nuclei/section in the i.c.v. Octreotide 24-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 9380276-18 1997 Intracerebroventricular sandostatin injection attenuated the GHRP-6-induced Fos response, from 53 +/- 6 nuclei/section in the i.c.v. growth hormone releasing hexapeptide 61-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 9365026-0 1997 Amphetamine sensitization augments amphetamine-induced Fos expression in the lateral habenula. Amphetamine 35-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 9365026-2 1997 To investigate the neuroanatomical basis of this phenomenon, we examined the effects of AMPH sensitization on AMPH-induced Fos expression in 24 regions of the rat brain. Amphetamine 88-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-126 9365026-2 1997 To investigate the neuroanatomical basis of this phenomenon, we examined the effects of AMPH sensitization on AMPH-induced Fos expression in 24 regions of the rat brain. Amphetamine 110-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-126 9365026-5 1997 As measured by Fos immunohistochemistry, the AMPH sensitization procedure enhanced subsequent AMPH-induced Fos expression in only one structure, the medial part of the lateral habenula. Amphetamine 45-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-18 9365026-5 1997 As measured by Fos immunohistochemistry, the AMPH sensitization procedure enhanced subsequent AMPH-induced Fos expression in only one structure, the medial part of the lateral habenula. Amphetamine 45-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 9365026-5 1997 As measured by Fos immunohistochemistry, the AMPH sensitization procedure enhanced subsequent AMPH-induced Fos expression in only one structure, the medial part of the lateral habenula. Amphetamine 94-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-18 9365026-5 1997 As measured by Fos immunohistochemistry, the AMPH sensitization procedure enhanced subsequent AMPH-induced Fos expression in only one structure, the medial part of the lateral habenula. Amphetamine 94-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 9359591-0 1997 Formalin-evoked Fos expression in spinal cord is enhanced in morphine-tolerant rats. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 9359591-0 1997 Formalin-evoked Fos expression in spinal cord is enhanced in morphine-tolerant rats. Morphine 61-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 9359591-4 1997 Although there was no significant difference in flinching behavior between the morphine-tolerant and control groups, we recorded significantly increased total Fos-like immunoreactivity at the L4/5 and L2 segments both ipsilateral and contralateral to the site of formalin injection in the morphine-tolerant rats compared to the control rats. Morphine 79-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-162 9359591-4 1997 Although there was no significant difference in flinching behavior between the morphine-tolerant and control groups, we recorded significantly increased total Fos-like immunoreactivity at the L4/5 and L2 segments both ipsilateral and contralateral to the site of formalin injection in the morphine-tolerant rats compared to the control rats. Formaldehyde 263-271 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-162 9359591-4 1997 Although there was no significant difference in flinching behavior between the morphine-tolerant and control groups, we recorded significantly increased total Fos-like immunoreactivity at the L4/5 and L2 segments both ipsilateral and contralateral to the site of formalin injection in the morphine-tolerant rats compared to the control rats. Morphine 289-297 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-162 9313909-0 1997 Quipazine and light have similar effects on c-fos induction in the rat suprachiasmatic nucleus. Quipazine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 9298238-0 1997 Recovery of hypothalamic NMDA-induced c-fos expression following neonatal glutamate (MSG) lesions. N-Methylaspartate 25-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 9298238-0 1997 Recovery of hypothalamic NMDA-induced c-fos expression following neonatal glutamate (MSG) lesions. Glutamic Acid 74-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 9298238-4 1997 Groups of neonatal (postnatal day (PD) 2) pups were injected with MSG, then stimulated on subsequent days (PD 3-29) with NMDA, known to induce c-fos expression in ARC. N-Methylaspartate 121-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-148 9298238-14 1997 We conclude that the recovery of hypothalamic function (i.e., onset of puberty) after a neonatal MSG lesion is coincident with the reappearance of a normal pattern of c-fos expression in response to NMDA stimulation. N-Methylaspartate 199-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 167-172 9298239-1 1997 Maternal treatment with cocaine or the D1-dopamine receptor agonist, SKF 38393, induces expression of the immediate-early gene, c-fos, in fetal rodent brain. Cocaine 24-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-133 9283705-15 1997 Both alpha, beta-meATP and bFGF rapidly and transiently induced the nuclear accumulation of Fos and Jun. , beta-meatp 10-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 9283705-16 1997 Both c-fos and c-jun induction by the purine analogue could be fully prevented by pretreatment with suramin. purine 38-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 5-10 9283705-16 1997 Both c-fos and c-jun induction by the purine analogue could be fully prevented by pretreatment with suramin. Suramin 100-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 5-10 9242432-11 1997 These inhibitors decreased crocidolite-induced c-fos but not c-jun levels, suggesting that tyrosine kinases have different regulatory roles in asbestos-induced c-fos versus c-jun signaling pathways. Asbestos, Crocidolite 27-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 9242432-11 1997 These inhibitors decreased crocidolite-induced c-fos but not c-jun levels, suggesting that tyrosine kinases have different regulatory roles in asbestos-induced c-fos versus c-jun signaling pathways. Asbestos, Crocidolite 27-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 9284361-14 1997 Taken together with the results recorded after intrathecal injection of naloxone-methiodide in tolerant rats, we conclude that the pattern of lumbar spinal cord Fos expression following systemic withdrawal is primarily a consequence of increased activity in opioid receptor-containing circuits intrinsic to the dorsal horn and that the magnitude of Fos expression is normally dampened by supraspinal and primary afferent-derived inhibitory inputs. N-methylnaloxone 72-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-164 9242432-0 1997 Inhibition of protein kinase C prevents asbestos-induced c-fos and c-jun proto-oncogene expression in mesothelial cells. Asbestos 40-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 9242432-1 1997 Asbestos and the phorbol ester tumor promoter, 12-O-tetradecanoylphorbol-13-acetate (TPA), increase c-fos and c-jun mRNA levels and AP-1 DNA binding activity in rat pleural mesothelial (RPM) cells, a target cell of asbestos-induced mesotheliomas (N. H. Heintz et al., Proc. Asbestos 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 9242432-1 1997 Asbestos and the phorbol ester tumor promoter, 12-O-tetradecanoylphorbol-13-acetate (TPA), increase c-fos and c-jun mRNA levels and AP-1 DNA binding activity in rat pleural mesothelial (RPM) cells, a target cell of asbestos-induced mesotheliomas (N. H. Heintz et al., Proc. Phorbol Esters 17-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 9242432-1 1997 Asbestos and the phorbol ester tumor promoter, 12-O-tetradecanoylphorbol-13-acetate (TPA), increase c-fos and c-jun mRNA levels and AP-1 DNA binding activity in rat pleural mesothelial (RPM) cells, a target cell of asbestos-induced mesotheliomas (N. H. Heintz et al., Proc. Tetradecanoylphorbol Acetate 47-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 9242432-1 1997 Asbestos and the phorbol ester tumor promoter, 12-O-tetradecanoylphorbol-13-acetate (TPA), increase c-fos and c-jun mRNA levels and AP-1 DNA binding activity in rat pleural mesothelial (RPM) cells, a target cell of asbestos-induced mesotheliomas (N. H. Heintz et al., Proc. Tetradecanoylphorbol Acetate 85-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 9242432-9 1997 Quantitation of Northern blots showed that fiber-associated c-fos/c-jun mRNA levels were significantly lower either after PKC alpha down-modulation or pretreatment with calphostin C. calphostin C 169-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 9313909-1 1997 The effects of the serotonin agonist, quipazine, on the induction of c-fos in the suprachiasmatic nucleus of the rat was examined at different times of the 24 h cycle. Quipazine 38-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 9313909-2 1997 Quipazine administered at night induced Fos production in a dose dependent manner (1, 3, 10, 30 mumol/kg) in the ventrolateral portion of the suprachiasmatic nucleus at ZT18. Quipazine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 9313909-4 1997 When compared to the effects of light pulses (2 lux/1 min), quipazine only caused c-fos induction at times when light caused induction. Quipazine 60-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 9231766-10 1997 Finally, the defective adipogenesis seen in epididymal preadipocytes from castrated rats was associated with reduced Fos protein induction in these cells, an alteration which was partly corrected by testosterone-treatment. Testosterone 199-211 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 9295196-8 1997 In the three brainstem structures the number of c-Fos-positive cells was elevated 8-10-fold in the clonidine-atipamezole group compared to the other groups. Clonidine 99-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 9308019-1 1997 The presence of Fos-labeled neurons at ultrastructural level was confirmed in the spinal superficial laminae following an injection of formalin into rat hindpaw in the present study. Formaldehyde 135-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 9295196-8 1997 In the three brainstem structures the number of c-Fos-positive cells was elevated 8-10-fold in the clonidine-atipamezole group compared to the other groups. atipamezole 109-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 9295196-10 1997 An increased number of c-Fos-positive neurons was also noted in the dorsal horn and intermediate layers of the thoracic spinal cord in the clonidine-atipamezole group compared to a sham-operated atipamezole-injected group. Clonidine 139-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 9295196-10 1997 An increased number of c-Fos-positive neurons was also noted in the dorsal horn and intermediate layers of the thoracic spinal cord in the clonidine-atipamezole group compared to a sham-operated atipamezole-injected group. atipamezole 149-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 9295196-10 1997 An increased number of c-Fos-positive neurons was also noted in the dorsal horn and intermediate layers of the thoracic spinal cord in the clonidine-atipamezole group compared to a sham-operated atipamezole-injected group. atipamezole 195-206 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 9295196-11 1997 In the RVL, 59% of c-Fos-positive cells contained alpha2A-adrenergic receptor-like immunoreactivity in clonidine-atipamezole treated (withdrawing) rats. Clonidine 103-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 9295196-11 1997 In the RVL, 59% of c-Fos-positive cells contained alpha2A-adrenergic receptor-like immunoreactivity in clonidine-atipamezole treated (withdrawing) rats. atipamezole 113-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 9295196-12 1997 In addition, one-third of the tyrosine hydroxylase (TH)-immunopositive cells in RVL were also c-Fos-positive in clonidine withdrawing rats where no TH-positive cells were also c-Fos-positive in RVL of control groups. Clonidine 112-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 9295196-17 1997 In conclusion, administration of the selective alpha2-antagonist atipamezole to rats chronically treated with the alpha2-adrenergic agonist clonidine triggers a powerful withdrawal syndrome associated with massive CNS expression of c-Fos protein. atipamezole 65-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 232-237 9295196-17 1997 In conclusion, administration of the selective alpha2-antagonist atipamezole to rats chronically treated with the alpha2-adrenergic agonist clonidine triggers a powerful withdrawal syndrome associated with massive CNS expression of c-Fos protein. Clonidine 140-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 232-237 9295199-8 1997 In contrast, DHTP did not affect PPE hnRNA, but inhibited the c-fos mRNA response to novelty. dihydrotestosterone propionate 13-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 9219975-5 1997 Several brain regions (including components of the lamina terminalis, the paraventricular and supraoptic nuclei of the hypothalamus, and the area postrema) were activated to express c-Fos similarly in adult and two-day-old rats after 2 M NaCl injection, consistent with previous reports implicating a subset of these regions in osmotically-stimulated drinking and neurohypophyseal secretion. Sodium Chloride 238-242 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 182-187 9219975-6 1997 In contrast, other areas of the brain that were activated to express c-Fos in adult rats after 2 M NaCl injection were not activated in neonates: these areas included the central nucleus of the amygdala, the parabrachial nucleus and catecholamine cell groups within the caudal medulla. Sodium Chloride 99-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 9214544-5 1997 Vc neurons in which c-fos protein-like immunoreactivity (Fos-LI) was induced by subcutaneous injection of formalin into the lip were considered nociceptive. Formaldehyde 106-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-25 9214544-5 1997 Vc neurons in which c-fos protein-like immunoreactivity (Fos-LI) was induced by subcutaneous injection of formalin into the lip were considered nociceptive. Formaldehyde 106-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 9214544-8 1997 Confocal laser-scanning microscopy revealed that axonal varicosities with 5-HT-LI were in close apposition to TMR-DA-labeled neurons showing Fos-LI in lamina I and the outer part of lamina II (lamina IIo), and that both axonal varicosities with 5-HT-LI and those binding I-B4 were in close apposition to single neuronal profiles labeled with TMR-DA. amsonic acid 114-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 9295196-0 1997 Atipamezole-precipitated clonidine withdrawal induces c-Fos expression in rat central nervous system. atipamezole 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 9295196-0 1997 Atipamezole-precipitated clonidine withdrawal induces c-Fos expression in rat central nervous system. Clonidine 25-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 9295196-5 1997 The brains of the clonidine-atipamezole group showed massive c-Fos expression (especially in di- and telencephalon) while the other groups showed either background levels of c-Fos-immunopositive cells (saline-saline and clonidine-saline groups) or a slight increase over background in selected areas (saline-atipamezole group). Clonidine 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 9295196-5 1997 The brains of the clonidine-atipamezole group showed massive c-Fos expression (especially in di- and telencephalon) while the other groups showed either background levels of c-Fos-immunopositive cells (saline-saline and clonidine-saline groups) or a slight increase over background in selected areas (saline-atipamezole group). atipamezole 28-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 9258997-4 1997 Fos-like immunoreactivity (Fos-in used as a marker of cellular activation) was induced by BK + captopril in regions of the brain previously associated with action of angiotensin (Ang) II, including the circumventricular organs and the magnocellular hypothalamic nuclei. bk + captopril 90-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9258997-4 1997 Fos-like immunoreactivity (Fos-in used as a marker of cellular activation) was induced by BK + captopril in regions of the brain previously associated with action of angiotensin (Ang) II, including the circumventricular organs and the magnocellular hypothalamic nuclei. bk + captopril 90-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 9258997-10 1997 Further, Fos-IR induced by BK + captopril was only partly (31%) reduced in the supraoptic and paraventricular nuclei of lesioned rats compared with sham operated controls. Captopril 32-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 9812870-0 1997 [Expression of c-fos in spinal cord, medulla oblongata and thalamus following epicardial application of adenosine]. Adenosine 104-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 9812870-1 1997 Effects of epicardial application of adenosine on the expression of c-fos proto-oncogene in spinal cord, medulla oblongata and thalamus were examined in 12 sinoaortic denervated and vagotomized anesthetized rats. Adenosine 37-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 9274930-0 1997 Anxiolytic homophthalazines increase Fos-like immunoreactivity in selected brain areas of the rat. homophthalazines 11-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 9274930-1 1997 Nerisopam, an anxiolytic and antipsychotic homophthalazine induces rapid, intense expression of Fos-like immunoreactivity in the rostral, dorsomedial and lateral parts of the striatum in the rat. homophthalazine 43-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-99 9208992-5 1997 At the PB level, c-Fos was expressed preferentially contralaterally, increasing with the applied temperatures in a dependent manner in the noxious range (r = 0.971, n = 25). pladienolide B 7-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 9218464-7 1997 Furthermore, staurosporine induced immediate-early genes including Nur77 and fos, but not jun. Staurosporine 13-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-80 9262162-1 1997 Prior 2-deoxyglucose and c-fos studies have demonstrated increased metabolic activity in a rostral dorsomedial area of the olfactory bulb in response to the vapor of propionic acid. propionic acid 166-180 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 9272839-0 1997 Desensitization of Fos protein induction in rat striatum and nucleus accumbens following repeated administration of delta9-tetrahydrocannabinol. Dronabinol 116-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 9272839-1 1997 The purpose of the present study was to clarify the effect of repeated administration of delta9-tetrahydrocannabinol (THC) on Fos protein induction in the rat brain. Dronabinol 89-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-129 9272839-1 1997 The purpose of the present study was to clarify the effect of repeated administration of delta9-tetrahydrocannabinol (THC) on Fos protein induction in the rat brain. Dronabinol 118-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-129 9272839-3 1997 attenuated the effect of THC (10 mg/kg) to induce the expression of Fos protein in rat striatum and nucleus accumbens. Dronabinol 25-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 9272839-4 1997 This desensitization of Fos protein induction might explain the rapid development of behavioral tolerance to repeated administration of THC. Dronabinol 136-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 9208992-9 1997 We conclude that, under our experimental procedures, noxious heat-induced c-Fos expression at the PB level depends on the intensity of the noxious stimulation. pladienolide B 98-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 9500663-4 1997 Administration of the excitatory amino acid (EAA) antagonist MK-801 (3 mg/kg) failed to inhibit either the acute or entraining effects of light on melatonin production and only partially (approximately 30%) prevented the induction of c-Fos in the SCN. Dizocilpine Maleate 61-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 234-239 9500663-6 1997 When the non-specific serotonin agonist quipazine was administered at CT 18, it mimicked both the acute and phase delaying effects of light on melatonin secretion and induced c-Fos in the SCN with a regional distribution identical to that observed following light treatment. Quipazine 40-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 175-180 9500663-8 1997 DOI (0.5 mg/kg) also induced c-Fos in the SCN and the induction was prevented by ritanserin and ketanserin. Ritanserin 81-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 9500663-8 1997 DOI (0.5 mg/kg) also induced c-Fos in the SCN and the induction was prevented by ritanserin and ketanserin. Ketanserin 96-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 9192514-4 1997 Phorbol myristate acetate (PMA) elicits a similar increase in c-fos and c-jun mRNAs, but is unable to stimulate transcription of collagenase in these cells. Tetradecanoylphorbol Acetate 0-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 9192514-4 1997 Phorbol myristate acetate (PMA) elicits a similar increase in c-fos and c-jun mRNAs, but is unable to stimulate transcription of collagenase in these cells. Tetradecanoylphorbol Acetate 27-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 9194045-6 1997 AP-1 constituent proteins were differentially susceptible to lithium, as cJun was reduced by 55%, cFos was unaffected by lithium, and an intermediate effect was observed with Jun B. Lithium 61-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-102 9221899-2 1997 We investigated the expression of the Fos family proteins following injection of the NMDA receptor agonist quinolinic acid (QA) into the rat striatum. Quinolinic Acid 107-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 9221899-2 1997 We investigated the expression of the Fos family proteins following injection of the NMDA receptor agonist quinolinic acid (QA) into the rat striatum. Quinolinic Acid 124-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 9202210-4 1997 Treatment of ovariectomized rats with estradiol and progesterone reversed the promoting effect of ovariectomy on proliferation, differentiation, and c-fos induction in perirenal preadipocytes, but not the MAP kinase activation observed during the proliferative phase. Estradiol 38-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 149-154 9202210-4 1997 Treatment of ovariectomized rats with estradiol and progesterone reversed the promoting effect of ovariectomy on proliferation, differentiation, and c-fos induction in perirenal preadipocytes, but not the MAP kinase activation observed during the proliferative phase. Progesterone 52-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 149-154 26735788-0 1997 Induction of Fos protein by 3,4- methylenedioxymethamphetamine (Ecstasy) in rat brain: regional differences in pharmacological manipulation. N-Methyl-3,4-methylenedioxyamphetamine 28-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 26735788-0 1997 Induction of Fos protein by 3,4- methylenedioxymethamphetamine (Ecstasy) in rat brain: regional differences in pharmacological manipulation. N-Methyl-3,4-methylenedioxyamphetamine 64-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 26735788-2 1997 In the present study, immunohistochemical techniques were used to assess the pattern of Fos protein produced by 3,4-methylenedioxymethamphetamine (MDMA) in several brain regions. N-Methyl-3,4-methylenedioxyamphetamine 112-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 26735788-2 1997 In the present study, immunohistochemical techniques were used to assess the pattern of Fos protein produced by 3,4-methylenedioxymethamphetamine (MDMA) in several brain regions. N-Methyl-3,4-methylenedioxyamphetamine 147-151 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 26735788-3 1997 Furthermore, we also studied the role of the dopamine D and D receptors and the N-methyl- D-aspartate (NMDA) receptor in the induction of Fos protein by MDMA. N-Methyl-3,4-methylenedioxyamphetamine 153-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-141 26735788-4 1997 A single administration of MDMA (5, 10 or 20 mg/kg) caused marked induction of Fos-immunoreactivity in several regions including frontal cortex, striatum and olfactory tubercle of rat brain, in a dose-dependent manner. N-Methyl-3,4-methylenedioxyamphetamine 27-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 26735788-6 1997 Furthermore, the induction of Fos protein in the striatum and olfactory tubercle after administration of MDMA (10 mg/kg) was blocked by pre-treatment with the dopamine D receptor antagonist SCH 23390 (1 mg/kg) or the NMDA receptor antagonist dizocilpine (1 mg/kg), but not by the dopamine D receptor antagonist (-)-sulpiride (100 mg/kg). N-Methyl-3,4-methylenedioxyamphetamine 105-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 26735788-6 1997 Furthermore, the induction of Fos protein in the striatum and olfactory tubercle after administration of MDMA (10 mg/kg) was blocked by pre-treatment with the dopamine D receptor antagonist SCH 23390 (1 mg/kg) or the NMDA receptor antagonist dizocilpine (1 mg/kg), but not by the dopamine D receptor antagonist (-)-sulpiride (100 mg/kg). Dizocilpine Maleate 242-253 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 26735788-6 1997 Furthermore, the induction of Fos protein in the striatum and olfactory tubercle after administration of MDMA (10 mg/kg) was blocked by pre-treatment with the dopamine D receptor antagonist SCH 23390 (1 mg/kg) or the NMDA receptor antagonist dizocilpine (1 mg/kg), but not by the dopamine D receptor antagonist (-)-sulpiride (100 mg/kg). Sulpiride 311-324 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 26735788-7 1997 However, the induction of Fos protein in the frontal cortex and hippocampus by MDMA was unaltered by pretreatment with SCH 23390 (1 mg/kg) or (-)-sulpiride (100 mg/kg). N-Methyl-3,4-methylenedioxyamphetamine 79-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 26735788-8 1997 These results suggest that MDMA induces the expression of Fos protein in several regions of rat brain, and that the expression of Fos protein by MDMA in the striatum and olfactory tubercle appears to be mediated at least in part by the dopamine D and NMDA receptors. N-Methyl-3,4-methylenedioxyamphetamine 27-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 26735788-8 1997 These results suggest that MDMA induces the expression of Fos protein in several regions of rat brain, and that the expression of Fos protein by MDMA in the striatum and olfactory tubercle appears to be mediated at least in part by the dopamine D and NMDA receptors. Dopamine 236-244 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-133 9262200-4 1997 Fos expression was prevented by pretreatment with the non-competitive N-methyl-D-aspartate (NMDA) glutamate receptor antagonist MK-801 (0.1 mg/kg) or the D1 dopamine receptor antagonist SCH-23390 (0.1 mg/kg), but not by pretreatment with the D2 receptor antagonist eticlopride (0.5 mg/kg). Dizocilpine Maleate 128-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9262200-4 1997 Fos expression was prevented by pretreatment with the non-competitive N-methyl-D-aspartate (NMDA) glutamate receptor antagonist MK-801 (0.1 mg/kg) or the D1 dopamine receptor antagonist SCH-23390 (0.1 mg/kg), but not by pretreatment with the D2 receptor antagonist eticlopride (0.5 mg/kg). SCH 23390 186-195 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9262200-4 1997 Fos expression was prevented by pretreatment with the non-competitive N-methyl-D-aspartate (NMDA) glutamate receptor antagonist MK-801 (0.1 mg/kg) or the D1 dopamine receptor antagonist SCH-23390 (0.1 mg/kg), but not by pretreatment with the D2 receptor antagonist eticlopride (0.5 mg/kg). eticlopride 265-276 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9262200-5 1997 Thirty-six hours after 6-hydroxydopamine lesion, a considerable reduction in treadmill-induced Fos expression was observed in both sides; however, Fos expression in the lesioned striatum was higher than in the contralateral intact striatum. Oxidopamine 23-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 9262200-6 1997 Several weeks after unilateral 6-hydroxydopamine lesion of the nigrostriatal system, treadmill-induced Fos expression was significantly, but not totally, reduced in the lesioned striatum. Oxidopamine 31-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-106 9194047-3 1997 As was the case for the two reference antipsychotics, the two benzamides enhanced c-Fos immunoreactivity in a number of forebrain regions. Benzamides 62-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 9211355-3 1997 mRNA levels of c-fos and egr-1 were markedly increased in kidneys after 90 minutes of standard perfusion with Krebs-Henseleit buffer containing albumin. Krebs-Henseleit solution 110-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 9194047-7 1997 Clozapine increased the number of Fos-like immunoreactive cells in the lateral septal nucleus and the diagonal band nucleus, but YM-43611, nemonapride, and haloperidol did not. Clozapine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 9195609-0 1997 Dopamine/glutamate interaction as studied by combining turning behaviour and c-Fos expression. Dopamine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 9195609-0 1997 Dopamine/glutamate interaction as studied by combining turning behaviour and c-Fos expression. Glutamic Acid 9-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 9195609-7 1997 Studies on c-fos expression induced by DA D1 agonists in the 6-OHDA lesioned striatum show that detection of Fos-like immunoreactivity correlates to the long-term but not the acute effects induced by DA receptor stimulation and NMDA receptor blockade. Oxidopamine 61-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 9211355-5 1997 Protective measures known to minimize morphological damage to the mTAL, including hyperoncotic perfusion, perfusion with glycine, or perfusion with a mixture of amino acids, decreased mRNA levels of c-fos and egr-1 in the outer medulla (by 50% and 35%, respectively) and the papilla (by 60 and 30%, respectively). Glycine 121-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-204 9195609-7 1997 Studies on c-fos expression induced by DA D1 agonists in the 6-OHDA lesioned striatum show that detection of Fos-like immunoreactivity correlates to the long-term but not the acute effects induced by DA receptor stimulation and NMDA receptor blockade. Oxidopamine 61-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-112 9211490-11 1997 The addition of tyrphostin, a specific tyrosine kinase inhibitor, prevented c-fos induction and inhibited HGF-induced [3H]thymidine incorporation. Tyrphostins 16-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 9258917-8 1997 In contrast, Fos-IR in the group given isotonic saline was lacking in these three brain regions. Sodium Chloride 48-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 9247076-5 1997 The polyclonal c-fos antibody was tested with Western blotting and the diffusion of 4-aminopyridine investigated with autoradiography of [3H]4-aminopyridine. [3h]4-aminopyridine 137-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 10072926-0 1997 Effect of tetrandrine on proto-oncogene c-fos expression in rat cerebrum. tetrandrine 10-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 10072926-1 1997 AIM: To detect the effect of tetrandrine (Tet) on c-fos gene expression in cerebrum induced by lindane, a neurotoxicant which activates Ca2+ channels. tetrandrine 29-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 10072926-1 1997 AIM: To detect the effect of tetrandrine (Tet) on c-fos gene expression in cerebrum induced by lindane, a neurotoxicant which activates Ca2+ channels. tet 42-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 10072926-1 1997 AIM: To detect the effect of tetrandrine (Tet) on c-fos gene expression in cerebrum induced by lindane, a neurotoxicant which activates Ca2+ channels. Hexachlorocyclohexane 95-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 10072926-6 1997 30 min prior to lindane reduced c-fos gene expression in a concentration-dependent manner. Hexachlorocyclohexane 16-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 10072926-8 1997 CONCLUSION: Tet inhibited c-fos gene expression in rat cerebrum induced by Ca2+ agonist-lindane. tet 12-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 10072926-8 1997 CONCLUSION: Tet inhibited c-fos gene expression in rat cerebrum induced by Ca2+ agonist-lindane. Hexachlorocyclohexane 88-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 9197270-4 1997 Withdrawal, induced by the cannabinoid antagonist SR 141716A, was accompanied by a marked elevation in extracellular CRF concentration and a distinct pattern of Fos activation in the central nucleus of the amygdala. Cannabinoids 27-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-164 9197270-4 1997 Withdrawal, induced by the cannabinoid antagonist SR 141716A, was accompanied by a marked elevation in extracellular CRF concentration and a distinct pattern of Fos activation in the central nucleus of the amygdala. Rimonabant 50-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-164 9218679-5 1997 With the exception of a 48-120 h period required for licking/biting behavior to be restored to its normal status, the suppressive effect on c-fos expression and other nociceptive behaviors disappeared 48 h following c-fos antisense oligodeoxynucleotide treatment. Oligodeoxyribonucleotides 232-252 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 216-221 9218679-0 1997 Intrathecally administered c-fos antisense oligodeoxynucleotide decreases formalin-induced nociceptive behavior in adult rats. Oligodeoxyribonucleotides 43-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 9218679-0 1997 Intrathecally administered c-fos antisense oligodeoxynucleotide decreases formalin-induced nociceptive behavior in adult rats. Formaldehyde 74-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 9218679-1 1997 c-fos antisense strategy was applied as a pharmacological approach to characterize its dose-dependent role and reversibility in the reduction of formalin-induced hyperalgesia. Formaldehyde 145-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9218679-5 1997 With the exception of a 48-120 h period required for licking/biting behavior to be restored to its normal status, the suppressive effect on c-fos expression and other nociceptive behaviors disappeared 48 h following c-fos antisense oligodeoxynucleotide treatment. Oligodeoxyribonucleotides 232-252 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 9218679-6 1997 The results suggest a pharmacological potential of c-fos antisense oligodeoxynucleotides in the central nervous system to block immediate-early genes and their resulting physiological consequence following noxious stimulus. Oligodeoxyribonucleotides 67-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 9191086-2 1997 Methamphetamine (n = 11) induced a marked increase in c-fos mRNA in the dorsomedial quadrant of the striatum and a 21% smaller, but still reliable, increase in the ventrolateral quadrant. Methamphetamine 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 9221946-7 1997 Nicotine significantly increased c-Fos expression in a dose-dependent manner in the brainstem regions examined. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 9219861-2 1997 Stimulation of adenosine A2A receptors by CGS 21680 (5 mg/kg) induced an increase in Fos-like immunoreactivity in the rat nucleus accumbens shell, while in the rostral pole and core CGS 21680 induced Fos-like immunoreactivity only after a high dose. cysteinylglycine 42-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 9219861-2 1997 Stimulation of adenosine A2A receptors by CGS 21680 (5 mg/kg) induced an increase in Fos-like immunoreactivity in the rat nucleus accumbens shell, while in the rostral pole and core CGS 21680 induced Fos-like immunoreactivity only after a high dose. cysteinylglycine 42-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 200-203 9219861-3 1997 CGS 21680 (5 mg/kg) stimulated c-fos expression also in the lateral septal nucleus and dorso-medial striatum, but not in the dorso-lateral striatum. cysteinylglycine 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 9219861-5 1997 Administration of the selective A2A antagonist SCH 58261 counteracted CGS 21680-induced Fos-like immunoreactivity. 5-amino-7-(2-phenylethyl)-2-(2-furyl)pyrazolo(4,3-e)-1,2,4-triazolo(1,5-c)pyrimidine 47-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 9219861-5 1997 Administration of the selective A2A antagonist SCH 58261 counteracted CGS 21680-induced Fos-like immunoreactivity. cysteinylglycine 70-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 9219861-6 1997 Lesions of the dopaminergic mesostriatal projection by 6-hydroxydopamine and stimulation of dopamine D2/D3 receptors by quinpirole, prevented CGS 21680-induced Fos-like immunoreactivity in the nucleus accumbens shell. Oxidopamine 55-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-163 9219861-6 1997 Lesions of the dopaminergic mesostriatal projection by 6-hydroxydopamine and stimulation of dopamine D2/D3 receptors by quinpirole, prevented CGS 21680-induced Fos-like immunoreactivity in the nucleus accumbens shell. Quinpirole 120-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-163 9219861-6 1997 Lesions of the dopaminergic mesostriatal projection by 6-hydroxydopamine and stimulation of dopamine D2/D3 receptors by quinpirole, prevented CGS 21680-induced Fos-like immunoreactivity in the nucleus accumbens shell. cysteinylglycine 142-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-163 9219875-4 1997 Tandospirone and ipsapirone each induced expression of Fos protein in the noradrenergic neurons of the locus coeruleus of conscious rats. tandospirone 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 9219875-4 1997 Tandospirone and ipsapirone each induced expression of Fos protein in the noradrenergic neurons of the locus coeruleus of conscious rats. ipsapirone 17-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 9178866-5 1997 In this study, examination of rotational behaviour and induction of Fos-like immunoreactivity were used to investigate changes in the striatal outflow systems in response to treatment with the D2 agonist quinpirole in 6-hydroxydopamine-lesioned rats that had been primed with apomorphine. Quinpirole 204-214 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 9178866-6 1997 Administration of apomorphine (0.5 mg/kg; three injections at three to six day intervals) permitted an otherwise inactive dose of quinpirole (0.25 mg/kg) to produce robust contralateral rotation and to induce the expression of Fos in striatal neurons belonging to the striato-nigro-entopeduncular ("direct") pathway. Apomorphine 18-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 227-230 9178866-6 1997 Administration of apomorphine (0.5 mg/kg; three injections at three to six day intervals) permitted an otherwise inactive dose of quinpirole (0.25 mg/kg) to produce robust contralateral rotation and to induce the expression of Fos in striatal neurons belonging to the striato-nigro-entopeduncular ("direct") pathway. Quinpirole 130-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 227-230 9178866-7 1997 The increase in contralateral rotation and ipsilateral striatal Fos expression following administration of quinpirole to apomorphine-primed rats was mediated by a D2-like receptor and did not appear to be due to a change in sensitivity of D2 receptors. Quinpirole 107-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 9178866-8 1997 Apomorphine priming also enhanced the ability of quinpirole to induce Fos expression in the globus pallidus, a target of the striatopallidal ("indirect") pathway. Apomorphine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 9178866-8 1997 Apomorphine priming also enhanced the ability of quinpirole to induce Fos expression in the globus pallidus, a target of the striatopallidal ("indirect") pathway. Quinpirole 49-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 9178866-9 1997 Western blot analysis confirmed that treatment with quinpirole induced the expression of c-Fos protein with no change in the expression of 35-37,000 mol. Quinpirole 52-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 9178866-10 1997 wt Fos-related antigens in apomorphine-primed rats treated with water or quinpirole. Water 64-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 3-6 9178866-10 1997 wt Fos-related antigens in apomorphine-primed rats treated with water or quinpirole. Quinpirole 73-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 3-6 9178866-12 1997 Thus, the quinpirole-dependent induction of striatal Fos in apomorphine-primed 6-hydroxydopamine-lesioned rats represents a qualitative alteration in striatal outflow. Quinpirole 10-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 9178866-12 1997 Thus, the quinpirole-dependent induction of striatal Fos in apomorphine-primed 6-hydroxydopamine-lesioned rats represents a qualitative alteration in striatal outflow. Oxidopamine 79-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 9169474-8 1997 The consequence of PKC-dependent JNK inhibition was reflected in c-Jun and c-Fos mRNA induction following treatment with thapsigargin and Ang II. Thapsigargin 121-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 9169474-9 1997 Thapsigargin, which only minimally induced c-Fos, produced a much greater and more prolonged c-Jun response than Ang II. Thapsigargin 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 26972126-0 1997 On the unique profile of action of clozapine as assessed with fos-protein induction in rat brain regions. Clozapine 35-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 26972126-6 1997 In this communication we present data on the in vivo profile of clozapine as revealed with Fos-protein expression. Clozapine 64-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-94 26972126-9 1997 Recent studies have shown that the antipsychotics haloperidol (Haldol ) and clozapine, when given acutely, induce different patterns of Fos-like immunoreactivity in the forebrain of the rat. Haloperidol 50-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-139 26972126-9 1997 Recent studies have shown that the antipsychotics haloperidol (Haldol ) and clozapine, when given acutely, induce different patterns of Fos-like immunoreactivity in the forebrain of the rat. Haloperidol 63-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-139 26972126-9 1997 Recent studies have shown that the antipsychotics haloperidol (Haldol ) and clozapine, when given acutely, induce different patterns of Fos-like immunoreactivity in the forebrain of the rat. Clozapine 76-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-139 9191092-0 1997 Induction of FOS and JUN proteins during focal epilepsy: congruences with and differences to [14C]deoxyglucose metabolism. Carbon-14 94-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 9131632-0 1997 Expression of Fos-like proteins in the preoptic area and hypothalamus of the rat brain following intracerebral or peripheral administration of colchicine. Colchicine 143-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 9177865-3 1997 We also found that a single dose of PGE2 induces the expression of early-response genes (c-fos, c-jun, and egr-1) in bone marrow cells within these two types of bone. Dinoprostone 36-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 9180903-6 1997 Submaximal doses of adenosine (10 nM-10 microM) were able to induce c-Fos expression in FRTL-5 cells. Adenosine 20-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 9185668-0 1997 Effect of a serotonin agonist (sumatriptan) on the peptidergic innervation of the rat cerebral dura mater and on the expression of c-fos in the caudal trigeminal nucleus in an experimental migraine model. Sumatriptan 31-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-136 9185668-6 1997 In the caudal trigeminal nucleus electrical stimulation of the trigeminal ganglion induces, in interneurons, increased expression of the oncoprotein c-fos which is not prevented by intravenous application of sumatriptan. Sumatriptan 208-219 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 149-154 9165496-1 1997 The mechanisms underlying the therapeutic efficacy of lithium in affective disorders are poorly understood; however, previous studies have established an influence of lithium on receptor-coupled and postreceptor signal transduction mechanisms, including the transcription factor c-fos. Lithium 54-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 279-284 9165496-2 1997 We investigated the effect of chronic lithium on basal, stress-, muscarinic-, and haloperidol-induced c-fos mRNA expression in various rat brain regions. Lithium 38-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 9165496-2 1997 We investigated the effect of chronic lithium on basal, stress-, muscarinic-, and haloperidol-induced c-fos mRNA expression in various rat brain regions. Haloperidol 82-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 9165496-3 1997 Chronic lithium produced significant reductions in basal c-fos expression in the frontal cortex and hippocampus, confirming our previous report. Lithium 8-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 9165496-5 1997 Pilocarpine-induced c-fos was significantly reduced in the frontal cortex and hippocampus by chronic lithium, but was enhanced in the occipital cortex and hypothalamus. Pilocarpine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 9165496-5 1997 Pilocarpine-induced c-fos was significantly reduced in the frontal cortex and hippocampus by chronic lithium, but was enhanced in the occipital cortex and hypothalamus. Lithium 101-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 9165496-6 1997 Haloperidol-induced c-fos was augmented in the striatum and pituitary, but reduced in the frontal cortex by chronic lithium treatment. Haloperidol 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 9165496-6 1997 Haloperidol-induced c-fos was augmented in the striatum and pituitary, but reduced in the frontal cortex by chronic lithium treatment. Lithium 116-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 9165496-7 1997 In regions in which haloperidol did not induce fos expression in control animals, fos levels after haloperidol were reduced after chronic lithium. Haloperidol 20-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 9165496-7 1997 In regions in which haloperidol did not induce fos expression in control animals, fos levels after haloperidol were reduced after chronic lithium. Haloperidol 99-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 9165496-7 1997 In regions in which haloperidol did not induce fos expression in control animals, fos levels after haloperidol were reduced after chronic lithium. Lithium 138-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 9165496-8 1997 One week after discontinuation of the lithium treatment, basal c-fos levels remained significantly lower in the frontal cortex and hippocampus, whereas the effects of stress, pilocarpine, or haloperidol on fos were normalized in most regions, except in the hippocampus, where the attenuated fos response to injection stress persisted. Lithium 38-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 9165496-8 1997 One week after discontinuation of the lithium treatment, basal c-fos levels remained significantly lower in the frontal cortex and hippocampus, whereas the effects of stress, pilocarpine, or haloperidol on fos were normalized in most regions, except in the hippocampus, where the attenuated fos response to injection stress persisted. Lithium 38-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 9165496-8 1997 One week after discontinuation of the lithium treatment, basal c-fos levels remained significantly lower in the frontal cortex and hippocampus, whereas the effects of stress, pilocarpine, or haloperidol on fos were normalized in most regions, except in the hippocampus, where the attenuated fos response to injection stress persisted. Haloperidol 191-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 206-209 9165496-9 1997 We suggest that repression of basal fos expression and inhibition and activation of inducible fos may be factors to be considered in the longer-term effects of lithium, leading to changes in expression of genes that regulate fos and are regulated by Fos, and ultimately to alterations in the functional activity of neural systems involved in the pathophysiology of affective disorder. Lithium 160-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-39 9165496-9 1997 We suggest that repression of basal fos expression and inhibition and activation of inducible fos may be factors to be considered in the longer-term effects of lithium, leading to changes in expression of genes that regulate fos and are regulated by Fos, and ultimately to alterations in the functional activity of neural systems involved in the pathophysiology of affective disorder. Lithium 160-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 9165496-9 1997 We suggest that repression of basal fos expression and inhibition and activation of inducible fos may be factors to be considered in the longer-term effects of lithium, leading to changes in expression of genes that regulate fos and are regulated by Fos, and ultimately to alterations in the functional activity of neural systems involved in the pathophysiology of affective disorder. Lithium 160-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 9165496-9 1997 We suggest that repression of basal fos expression and inhibition and activation of inducible fos may be factors to be considered in the longer-term effects of lithium, leading to changes in expression of genes that regulate fos and are regulated by Fos, and ultimately to alterations in the functional activity of neural systems involved in the pathophysiology of affective disorder. Lithium 160-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 250-253 9133401-1 1997 The distribution of neurons in the medullary reticular formation (RF) activated by the ingestion of sucrose or rejection of quinine was examined using standard immunohistochemical techniques to detect the expression of the Fos protein product of the immediate-early gene c-fos. Sucrose 100-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 223-226 9187311-5 1997 PB withdrawal seizures were followed by increased expression of c-fos mRNA in the hippocampus and cerebral cortex and of c-jun mRNA in the cerebral cortex. Phenobarbital 0-2 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 9187311-6 1997 The induction of c-fos and c-jun mRNA was suppressed by administration of MK-801. Dizocilpine Maleate 74-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 9176333-2 1997 Immunocytochemical staining of Fos was used to identify spinal neurons that receive excitatory inputs from the DNP in anesthetized rats. dnp 111-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 9176333-4 1997 Dissection as well as stimulation of the DNP elicited a comparable increase in Fos staining. dnp 41-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 9176333-7 1997 Stimulation of the DNP elicited both increased Fos labeling and ICP after spinalization, demonstrating the presence of a supraspinal inhibitory control exerted on the polysynaptic intraspinal circuitry responsible for reflexive penile erection. dnp 19-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 9112413-9 1997 Estradiol stimulated c-fos expression in lactotrophs and folliculo-stellate cells within the anterior lobe without affecting either the intermediate or neural lobes. Estradiol 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 9112413-10 1997 In a second experiment, c-fos messenger RNA levels were measured by solution hybridization in anterior pituitaries and uteri from estradiol-treated rats. Estradiol 130-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 9141428-12 1997 Despite abrogation of hepatocyte PD changes, proto-oncogene mRNA and protein levels in saline- and GABA-treated rats were either similar or, in the case of c-fos and c-jun, increased five- to sevenfold in GABA-treated rats. gamma-Aminobutyric Acid 99-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 9174082-0 1997 Increased sodium appetite stimulates c-fos expression in the organum vasculosum of the lamina terminalis. Sodium 10-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 9174082-1 1997 The relation between c-fos expression in the forebrain of Lister hooded rats and water and NaCl intakes was examined in response to systemic injection of angiotensin II, desoxycorticosterone, angiotensin II and desoxycorticosterone together, frusemide or low sodium diet, all treatments that induce a sodium appetite. Water 81-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 9174082-1 1997 The relation between c-fos expression in the forebrain of Lister hooded rats and water and NaCl intakes was examined in response to systemic injection of angiotensin II, desoxycorticosterone, angiotensin II and desoxycorticosterone together, frusemide or low sodium diet, all treatments that induce a sodium appetite. Sodium Chloride 91-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 9174082-1 1997 The relation between c-fos expression in the forebrain of Lister hooded rats and water and NaCl intakes was examined in response to systemic injection of angiotensin II, desoxycorticosterone, angiotensin II and desoxycorticosterone together, frusemide or low sodium diet, all treatments that induce a sodium appetite. Desoxycorticosterone 170-190 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 9174082-1 1997 The relation between c-fos expression in the forebrain of Lister hooded rats and water and NaCl intakes was examined in response to systemic injection of angiotensin II, desoxycorticosterone, angiotensin II and desoxycorticosterone together, frusemide or low sodium diet, all treatments that induce a sodium appetite. Desoxycorticosterone 211-231 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 9174082-9 1997 Animals given angiotensin II following desoxycorticosterone pretreatment showed patterns of c-fos expression that did not differ from those of angiotensin II alone. Desoxycorticosterone 39-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 9174082-11 1997 Frusemide gave a similar pattern of staining to desoxycorticosterone, stimulating c-fos expression in the same regions but to a lesser extent. Furosemide 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 9174082-11 1997 Frusemide gave a similar pattern of staining to desoxycorticosterone, stimulating c-fos expression in the same regions but to a lesser extent. Desoxycorticosterone 48-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 9174082-12 1997 A low salt diet resulted in increased c-fos expression only in the organum vasculosum of the laminal terminalis. Salts 6-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 9174082-13 1997 Therefore, five different treatments that induced increased sodium appetite evoked distinct patterns of c-fos expression in the anteroventral region of the third ventricle of the rat forebrain. Sodium 60-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 9174082-14 1997 Since the common feature was induction of c-fos in the organum vasculosum of the laminal terminalis, these results suggest a key role for this structure in the development of increased sodium appetite. Sodium 185-191 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 9180213-0 1997 Protein synthesis inhibitor cycloheximide dose-dependently decreases formalin-induced c-Fos protein and behavioral hyperalgesia in rats. Cycloheximide 28-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 9180213-0 1997 Protein synthesis inhibitor cycloheximide dose-dependently decreases formalin-induced c-Fos protein and behavioral hyperalgesia in rats. Formaldehyde 69-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 9180213-1 1997 We had previously demonstrated that c-fos antisense oligodeoxynucleotides dose-dependently suppressed formalin-induced c-Fos protein and behavioral hyperalgesia. Oligodeoxyribonucleotides 52-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 9180213-1 1997 We had previously demonstrated that c-fos antisense oligodeoxynucleotides dose-dependently suppressed formalin-induced c-Fos protein and behavioral hyperalgesia. Oligodeoxyribonucleotides 52-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 9180213-1 1997 We had previously demonstrated that c-fos antisense oligodeoxynucleotides dose-dependently suppressed formalin-induced c-Fos protein and behavioral hyperalgesia. Formaldehyde 102-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 9180213-1 1997 We had previously demonstrated that c-fos antisense oligodeoxynucleotides dose-dependently suppressed formalin-induced c-Fos protein and behavioral hyperalgesia. Formaldehyde 102-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 9180213-2 1997 To test whether de novo protein synthesis is required for the development of persistent pain after peripheral inflammation, we observed formalin-induced spinal c-Fos protein and nociceptive behaviors following pretreatment with cycloheximide, a protein synthesis inhibitor. Formaldehyde 136-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 9180213-2 1997 To test whether de novo protein synthesis is required for the development of persistent pain after peripheral inflammation, we observed formalin-induced spinal c-Fos protein and nociceptive behaviors following pretreatment with cycloheximide, a protein synthesis inhibitor. Cycloheximide 228-241 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 9180213-3 1997 Cycloheximide dose-dependently inhibited formalin-induced spinal c-Fos protein and tonic nociceptive responses. Cycloheximide 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 9180213-3 1997 Cycloheximide dose-dependently inhibited formalin-induced spinal c-Fos protein and tonic nociceptive responses. Formaldehyde 41-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 9180214-4 1997 Two months after the administration of streptozotocin (STZ) to Wistar rats, the number of Fos-immunoreactive cells significantly decreased, although 1 week after the administration of STZ, the number had not yet changed in these STZ-induced diabetic rats. Streptozocin 39-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 9180214-4 1997 Two months after the administration of streptozotocin (STZ) to Wistar rats, the number of Fos-immunoreactive cells significantly decreased, although 1 week after the administration of STZ, the number had not yet changed in these STZ-induced diabetic rats. Streptozocin 55-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 9180221-0 1997 Induction of Fos-like immunoreactivity in the lower brainstem and the spinal cord of the rat by intraperitoneal administration of an endogenous satiety substance, 2-buten-4-olide. butenolide 163-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 9180221-1 1997 Induction of Fos in neurons by intraperitoneal injection of 2-buten-4-olide (2-B40), an endogenous satiety substance, was studied immunohistochemically in the brainstem and spinal cord of the rat. butenolide 60-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 9178855-2 1997 Fos expression in the suprachiasmatic nucleus (SCN) of rats induced by photic stimulation (300 lux, 1 h) during the early subjective night of the rats was inhibited by treatment with CMZ (10 mg/kg i.p.) calmidazolium 183-186 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9187343-0 1997 Ethanol-induced c-Fos expression in rat lines selected for low and high alcohol consumption. Ethanol 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 9187343-0 1997 Ethanol-induced c-Fos expression in rat lines selected for low and high alcohol consumption. Alcohols 72-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 9187343-3 1997 Although line differences were noted in several brain regions, the principal finding was that alcohol-nonpreferring lines (NP and ANA) displayed greater c-Fos expression in the locus coeruleus (LC) relative to the alcohol-preferring lines (P and AA) following injection of 3.0 g ethanol/kg. Alcohols 94-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-158 9131632-10 1997 These studies revealed negligible immunolabeling for Fos 24 hr after vehicle treatment, 24 hr after intracerebral delivery of colchicine, Fos-li was observed within the medial preoptic area, the arcuate nucleus, the supraoptic nucleus, and parvocellular neurons in the paraventricular nucleus. Colchicine 126-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-141 9131632-11 1997 Animals treated with colchicine by gavage exhibited Fos-immunopositive neurons in the same sites, but additional immunolabeling for Fos was also observed within the median preoptic nucleus, suprachiasmatic nucleus, dorsomedial nucleus, and magnocellular neurons in the paraventricular nucleus. Colchicine 21-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 9131632-11 1997 Animals treated with colchicine by gavage exhibited Fos-immunopositive neurons in the same sites, but additional immunolabeling for Fos was also observed within the median preoptic nucleus, suprachiasmatic nucleus, dorsomedial nucleus, and magnocellular neurons in the paraventricular nucleus. Colchicine 21-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-135 9131632-12 1997 These results suggest, first of all, that neuronal responses to colchicine exposure include the synthesis of Fos-like proteins in a number of brain sites, at least over the time frame examined here. Colchicine 64-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-112 9200179-0 1997 Chronic treatment with systemic morphine induced tolerance to the systemic and peripheral antinociceptive effects of morphine on both carrageenin induced mechanical hyperalgesia and spinal c-Fos expression in awake rats. Morphine 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 189-194 9200179-0 1997 Chronic treatment with systemic morphine induced tolerance to the systemic and peripheral antinociceptive effects of morphine on both carrageenin induced mechanical hyperalgesia and spinal c-Fos expression in awake rats. Morphine 117-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 189-194 9200179-3 1997 morphine were investigated on spinal c-Fos expression induced 2 h after intraplantar carrageenin (6 mg/150 microliters of saline) and on carrageenin (2 mg/150 microliters of saline) induced mechanical hyperalgesia, at day 4, in both naive and chronic morphine treated (80 mg/kg/day s.c. on days 1, 2 and 3) rats. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 9200179-6 1997 morphine significantly decreased spinal c-Fos expression (64 +/- 4% and 44 +/- 4% reduction of control carrageenin c-Fos expression, P < 0.0001 for both, respectively) and mechanical hyperalgesia (maximal increase: 326 +/- 29%, P < 0.0001 and 87 +/- 5%, P < 0.005 of control carrageenin paw pressure vocalisation threshold (VTPP), respectively), which only developed in the carrageenin injected paw. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 9200179-6 1997 morphine significantly decreased spinal c-Fos expression (64 +/- 4% and 44 +/- 4% reduction of control carrageenin c-Fos expression, P < 0.0001 for both, respectively) and mechanical hyperalgesia (maximal increase: 326 +/- 29%, P < 0.0001 and 87 +/- 5%, P < 0.005 of control carrageenin paw pressure vocalisation threshold (VTPP), respectively), which only developed in the carrageenin injected paw. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 9200179-10 1997 These studies based on spinal c-Fos expression as an indirect marker of spinal nociceptive processes and on behavioural experiments clearly revealed that chronic treatment with systemic morphine induced tolerance to both its systemic and peripheral effects. Morphine 186-194 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 9787241-1 1997 Immunoreactivity of the immediate early gene c-fos was used to investigate changes in the activity of brainstem neurons in response to acute stressors like immobilization, formalin-induced pain, cold exposure, hemorrhage and insulin-induced hypoglycemia. Formaldehyde 172-180 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 9134962-8 1997 Supershift analysis with specific antibodies against the members of Fos and Jun protein families (c-Fos, Fos B, c-Jun, Jun B, Jun D) revealed that the NMDA-induced AP-1 complex was composed predominantly of Jun D and c-Fos. N-Methylaspartate 151-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 9135101-0 1997 Dizocilpine maleate, an N-methyl-D-aspartate antagonist, inhibits dipsogenic responses and C-Fos expression induced by intracerebral infusion of angiotensin II. Dizocilpine Maleate 0-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 9135101-0 1997 Dizocilpine maleate, an N-methyl-D-aspartate antagonist, inhibits dipsogenic responses and C-Fos expression induced by intracerebral infusion of angiotensin II. N-Methylaspartate 24-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 9135101-6 1997 Dizocilpine also decreased the angiotensin II-evoked expression of c-fos in the median preoptic nucleus, supraoptic nucleus and the medial (parvicellular) and lateral (magnocellular) parts of the hypothalamic paraventricular nucleus, as well as in the nucleus of the solitary tract and the lateral parabrachial nucleus. Dizocilpine Maleate 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 9134962-8 1997 Supershift analysis with specific antibodies against the members of Fos and Jun protein families (c-Fos, Fos B, c-Jun, Jun B, Jun D) revealed that the NMDA-induced AP-1 complex was composed predominantly of Jun D and c-Fos. N-Methylaspartate 151-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 9134962-8 1997 Supershift analysis with specific antibodies against the members of Fos and Jun protein families (c-Fos, Fos B, c-Jun, Jun B, Jun D) revealed that the NMDA-induced AP-1 complex was composed predominantly of Jun D and c-Fos. N-Methylaspartate 151-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 217-222 9134962-10 1997 In contrast, application of AMPA plus cyclothiazide induced an AP-1 transcription with contribution of Jun D, c-Fos, Fos B, c-Jun and Jun B proteins. cyclothiazide 38-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 9145767-0 1997 Pretreatment with 5-HT1A receptor agonist flesinoxan attenuates Fos protein in rat hypothalamus. flesinoxan 42-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 9145767-3 1997 Male rats received two injections with either flesinoxan or vehicle within 24 h. Flesinoxan challenge enhanced Fos immunoreactivity in the paraventricular nucleus of the hypothalamus, the central amygdala, and the dorsolateral part of the bed nucleus of the stria terminalis and plasma corticosterone levels in the vehicle-pretreated rats. flesinoxan 46-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 9145767-3 1997 Male rats received two injections with either flesinoxan or vehicle within 24 h. Flesinoxan challenge enhanced Fos immunoreactivity in the paraventricular nucleus of the hypothalamus, the central amygdala, and the dorsolateral part of the bed nucleus of the stria terminalis and plasma corticosterone levels in the vehicle-pretreated rats. flesinoxan 81-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 9145767-4 1997 Flesinoxan pretreatment resulted in an attenuated response of plasma corticosterone levels and Fos-positive neurons in the paraventricular nucleus of the hypothalamus, but not in the central amygdala and the bed nucleus after a flesinoxan challenge. flesinoxan 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 9092609-9 1997 These observations suggest that age-dependent hippocampal dysfunction may be associated with a selective change in the dynamic activity of signaling pathways upstream of c-fos, possibly involving calcium regulation. Calcium 196-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-175 9159506-1 1997 Expression of proto-oncogene c-fos was immunohistochemically examined in the central and basolateral amygdaloid nuclei in rats after ingestion of taste solutions (0.5 M sucrose or 0.005 M saccharin), intragastric infusion of these solutions, or an intraperitoneal injection of malaise-inducing lithium chloride (LiCl). Saccharin 188-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-34 9159506-1 1997 Expression of proto-oncogene c-fos was immunohistochemically examined in the central and basolateral amygdaloid nuclei in rats after ingestion of taste solutions (0.5 M sucrose or 0.005 M saccharin), intragastric infusion of these solutions, or an intraperitoneal injection of malaise-inducing lithium chloride (LiCl). Lithium Chloride 294-310 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-34 9159506-1 1997 Expression of proto-oncogene c-fos was immunohistochemically examined in the central and basolateral amygdaloid nuclei in rats after ingestion of taste solutions (0.5 M sucrose or 0.005 M saccharin), intragastric infusion of these solutions, or an intraperitoneal injection of malaise-inducing lithium chloride (LiCl). Lithium Chloride 312-316 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-34 9159506-2 1997 C-Fos-like immunoreactive neurons were distributed most densely in the central nucleus in response to the LiCl injection, followed by the ingestion and intragastric infusion of sucrose. Lithium Chloride 106-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9159506-2 1997 C-Fos-like immunoreactive neurons were distributed most densely in the central nucleus in response to the LiCl injection, followed by the ingestion and intragastric infusion of sucrose. Sucrose 177-184 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9159506-3 1997 The intraoral infusion of sucrose, but not of saccharin, elicited intense c-fos expression in the central nucleus after establishment of conditioned taste aversion to these taste stimuli. Sucrose 26-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 9160980-0 1997 c-Fos induction in the nucleus of the solitary tract of sodium-depleted rats by salt intake, peripheral bombesin, and the combination. Sodium 56-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9160980-0 1997 c-Fos induction in the nucleus of the solitary tract of sodium-depleted rats by salt intake, peripheral bombesin, and the combination. Salts 80-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9160981-0 1997 Salt-loading induces decreased POMC mRNA levels, increased alpha-MSH immunoreactivity, and sustained elevated fos expression in rat pituitary intermediate lobe melanotropes. Salts 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 9163536-1 1997 Intraplantar injection of dilute formalin evokes brief (Phase 1) and persistent (Phase 2) increases in primary afferent activity, pain behavior, and cardiovascular responses, and induces spinal cord Fos-like immunoreactivity (Fos-LI). Formaldehyde 33-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-202 9163536-1 1997 Intraplantar injection of dilute formalin evokes brief (Phase 1) and persistent (Phase 2) increases in primary afferent activity, pain behavior, and cardiovascular responses, and induces spinal cord Fos-like immunoreactivity (Fos-LI). Formaldehyde 33-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 226-229 9163536-6 1997 Also, in capsaicin-treated rats, we counted 59% fewer Fos-labeled neurons in the spinal cord. Capsaicin 9-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 9125414-2 1997 (1) During postnatal development without stimulation, c-Fos is absent in DCN and VCN from postnatal day (P) 12 to adulthood, but shows a strong expression in the IC which declines during maturation. dcn 73-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 9109393-0 1997 Essential role of Rac GTPase in hydrogen peroxide-induced activation of c-fos serum response element. Hydrogen Peroxide 32-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 9109393-1 1997 In the present study, we investigated whether hydrogen peroxide activates c-fos serum response element (SRE) in Rat-2 fibroblast cells. Hydrogen Peroxide 46-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 9125437-3 1997 Additionally, Fos-labelled neurons were found in the lower thoracolumbar spinal cord predominantly ipsilateral to the side of ARN stimulation. arn 126-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 9108355-8 1997 Numerous cells in bone marrow (both in the tibia and calvaria) expressed high levels of c-fos in response to PGE2. Dinoprostone 109-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 9140695-7 1997 In both the cerebral cortex and the hippocampus the stress-induced increase in c-fos mRNA was inhibited by MK-801, suggesting that it is mediated via NMDA receptors. Dizocilpine Maleate 107-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 9140695-8 1997 In the hippocampus, propranolol had a similar effect, indicating that beta-adrenergic receptors are also involved in the stress-induced increase in c-fos mRNA. Propranolol 20-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 9140695-10 1997 On the other hand, the increase in c-fos mRNA produced by the stress of the injection was inhibited in the cerebral cortex by diazepam or propranolol and in the hippocampus only by diazepam. Diazepam 126-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 9140695-10 1997 On the other hand, the increase in c-fos mRNA produced by the stress of the injection was inhibited in the cerebral cortex by diazepam or propranolol and in the hippocampus only by diazepam. Propranolol 138-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 9140695-10 1997 On the other hand, the increase in c-fos mRNA produced by the stress of the injection was inhibited in the cerebral cortex by diazepam or propranolol and in the hippocampus only by diazepam. Diazepam 181-189 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 9140695-11 1997 Furthermore, administration of MK-801 resulted in an increase in c-fos mRNA in the hippocampus of the nonstressed animals. Dizocilpine Maleate 31-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 9168840-1 1997 Exposure of the rodent striatum to quinolinic acid (QA, N-methyl-D-aspartate receptor agonist) induces immediate early gene (IEG; c-fos, c-jun, jun-B, zif/268) expression that may extend 12-24 h after injection. Quinolinic Acid 35-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 9168840-1 1997 Exposure of the rodent striatum to quinolinic acid (QA, N-methyl-D-aspartate receptor agonist) induces immediate early gene (IEG; c-fos, c-jun, jun-B, zif/268) expression that may extend 12-24 h after injection. Quinolinic Acid 52-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 9168840-3 1997 As early as 30 min after intrastriatal injection, both saline and QA increased c-fos mRNA levels. Sodium Chloride 55-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 9147294-4 1997 In the present study we used hybridization histochemical methods to compare the effects of ADX on the induction over time of two independent IEG markers, c-fos and NGFI-B, in hypophysiotropic CRF neurons. Adenylyl sulfate 91-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 9147294-9 1997 In the chronically ADX rat, CRF neurons are capable of c-fos expression since animals subjected to an acute injection of hypertonic saline 5 days after ADX displayed a robust induction of Fos-ir in the parvocellular PVH. Sodium Chloride 132-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 9147294-9 1997 In the chronically ADX rat, CRF neurons are capable of c-fos expression since animals subjected to an acute injection of hypertonic saline 5 days after ADX displayed a robust induction of Fos-ir in the parvocellular PVH. Sodium Chloride 132-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 188-191 9103499-7 1997 CI988 (3 mg/kg) significantly and strongly reduced the number of carrageenin-induced, superficial, Fos-like-immunoreactive neurons (55 +/- 5% reduction of control carrageenin c-Fos expression, P < .0001). PD 134308 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 175-180 9103499-7 1997 CI988 (3 mg/kg) significantly and strongly reduced the number of carrageenin-induced, superficial, Fos-like-immunoreactive neurons (55 +/- 5% reduction of control carrageenin c-Fos expression, P < .0001). Carrageenan 65-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 9103499-7 1997 CI988 (3 mg/kg) significantly and strongly reduced the number of carrageenin-induced, superficial, Fos-like-immunoreactive neurons (55 +/- 5% reduction of control carrageenin c-Fos expression, P < .0001). Carrageenan 65-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 175-180 9103499-11 1997 These results show that RB101 dose-dependently decreases carrageenin-evoked spinal c-Fos expression. Carrageenan 57-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 9181492-0 1997 D1 dopamine receptor agonist (SKF-38393) induction of Fos immunoreactivity in progestin receptor-containing areas of female rat brain. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 30-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 9181492-3 1997 Based on these data, neuronal response to the D1 receptor agonist SKF-38393 was assessed by expression of the immediate early gene protein, Fos. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 66-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-143 9147381-2 1997 Consistent with previously published observations, paw formalin injection evoked a distinct pattern of FOS protein expression in L3-L5 spinal segments. Formaldehyde 55-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-106 9147381-5 1997 In contrast, ipsilateral paw formalin injection, if administered 4 h after carrageenan-induced knee inflammation, evoked significantly fewer FOS positive neurons in all laminar and segmental levels analyzed as compared with formalin injected animals but without previous knee joint inflammation. Formaldehyde 29-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 9178036-1 1997 Addition of lead acetate to PC 12 pheochromocytoma cells elicits induction of c-fos, an immediate early response gene. lead acetate 12-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 9178036-2 1997 Induction of c-fos was concentration- and time-dependent: the lowest concentration of lead acetate tested that induced c-fos was 10 microM; induction was observed after a 30 min incubation and remained high after 90 min. lead acetate 86-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 9178036-2 1997 Induction of c-fos was concentration- and time-dependent: the lowest concentration of lead acetate tested that induced c-fos was 10 microM; induction was observed after a 30 min incubation and remained high after 90 min. lead acetate 86-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 9178036-3 1997 Treatment with lead acetate and cycloheximide superinduced c-fos mRNA. lead acetate 15-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 9178036-3 1997 Treatment with lead acetate and cycloheximide superinduced c-fos mRNA. Cycloheximide 32-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 9178036-4 1997 Actinomycin D, an inhibitor of mRNA transcription, decreased the level of c-fos mRNA induced by lead acetate by almost 80%. Dactinomycin 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 9178036-4 1997 Actinomycin D, an inhibitor of mRNA transcription, decreased the level of c-fos mRNA induced by lead acetate by almost 80%. lead acetate 96-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 9065519-6 1997 In subsequent experiments, alcohol inhibited the surge of LHRH (measured by push-pull cannulation) and LHRH neuronal activation (measured by Fos labeling in LHRH neurons). Alcohols 27-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 9103499-0 1997 Antinociceptive effects of RB101, a complete inhibitor of enkephalin-catabolizing enzymes, are enhanced by a cholecystokinin type B receptor antagonist, as revealed by noxiously evoked spinal c-Fos expression in rats. RB 101 27-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 192-197 9103499-2 1997 Spinal c-Fos expression was observed 90 min after intraplantar carrageenin (6 mg/150 microl saline), with Fos-like-immunoreactive neurons preferentially located in the superficial laminae of the spinal dorsal horn. Carrageenan 63-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 7-12 9103499-2 1997 Spinal c-Fos expression was observed 90 min after intraplantar carrageenin (6 mg/150 microl saline), with Fos-like-immunoreactive neurons preferentially located in the superficial laminae of the spinal dorsal horn. Sodium Chloride 92-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 7-12 9103499-2 1997 Spinal c-Fos expression was observed 90 min after intraplantar carrageenin (6 mg/150 microl saline), with Fos-like-immunoreactive neurons preferentially located in the superficial laminae of the spinal dorsal horn. Sodium Chloride 92-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 9103499-3 1997 Intravenous RB101 (10, 20 and 40 mg/kg) dose-dependently reduced the number of superficial Fos-like-immunoreactive neurons (r2 = 0.739, P < .0001), with 63 +/- 2% (P < .0001) reduction for the highest dose. RB 101 12-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-94 9103499-7 1997 CI988 (3 mg/kg) significantly and strongly reduced the number of carrageenin-induced, superficial, Fos-like-immunoreactive neurons (55 +/- 5% reduction of control carrageenin c-Fos expression, P < .0001). PD 134308 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 9125414-4 1997 (2) Between P15 and P35 stimulation of rats with 8 kHz for 5 min at 70 dB SPL evoked expression of c-Fos in a moderate number of cells of DCN, VCN and IC. dcn 138-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 9125414-11 1997 When compared with acutely stimulated rats at P35, both repetitive acoustic stimulation protocols reduced the c-Fos immunoreactivity by 50%-75% in the DCN, VCN and IC. dcn 151-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 9125414-11 1997 When compared with acutely stimulated rats at P35, both repetitive acoustic stimulation protocols reduced the c-Fos immunoreactivity by 50%-75% in the DCN, VCN and IC. polyacrylonitrile 156-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 9097389-2 1997 Administration of electroconvulsive seizures (ECS), tranylcypromine, or imipramine, three different classes of antidepressants, increased the expression of c-Fos mRNA and immunoreactivity in rat frontal cortex, but the magnitude of the increase for each treatment differed and the effect of imipramine was preceded by inhibition of c-Fos expression. Tranylcypromine 52-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 9097389-2 1997 Administration of electroconvulsive seizures (ECS), tranylcypromine, or imipramine, three different classes of antidepressants, increased the expression of c-Fos mRNA and immunoreactivity in rat frontal cortex, but the magnitude of the increase for each treatment differed and the effect of imipramine was preceded by inhibition of c-Fos expression. Tranylcypromine 52-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 332-337 9097389-2 1997 Administration of electroconvulsive seizures (ECS), tranylcypromine, or imipramine, three different classes of antidepressants, increased the expression of c-Fos mRNA and immunoreactivity in rat frontal cortex, but the magnitude of the increase for each treatment differed and the effect of imipramine was preceded by inhibition of c-Fos expression. Imipramine 72-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 9097389-2 1997 Administration of electroconvulsive seizures (ECS), tranylcypromine, or imipramine, three different classes of antidepressants, increased the expression of c-Fos mRNA and immunoreactivity in rat frontal cortex, but the magnitude of the increase for each treatment differed and the effect of imipramine was preceded by inhibition of c-Fos expression. Imipramine 72-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 332-337 9097389-2 1997 Administration of electroconvulsive seizures (ECS), tranylcypromine, or imipramine, three different classes of antidepressants, increased the expression of c-Fos mRNA and immunoreactivity in rat frontal cortex, but the magnitude of the increase for each treatment differed and the effect of imipramine was preceded by inhibition of c-Fos expression. Imipramine 291-301 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 9097389-5 1997 ECS- and tranylcypromine induction of c-Fos immunoreactivity in frontal cortex was partially inhibited by pretreatment with specific antagonists for alpha 1-adrenergic, beta-adrenergic, and 5-HT2A/2C, but not D2-dopamine receptors. Tranylcypromine 9-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 9097389-6 1997 ECS induction of c-Fos was also inhibited by D1-dopamine and NMDA glutamate receptor antagonists, suggesting that the greater induction of c-Fos by ECS results from activation of these, and possibly other, neurotransmitter receptors. d1-dopamine 45-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 9097389-6 1997 ECS induction of c-Fos was also inhibited by D1-dopamine and NMDA glutamate receptor antagonists, suggesting that the greater induction of c-Fos by ECS results from activation of these, and possibly other, neurotransmitter receptors. d1-dopamine 45-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 9097389-9 1997 The results demonstrate that ECS- and tranylcypromine induction of c-Fos is mediated by activation of several different neurotransmitter receptors, but that the exact pharmacological profile is different for each treatment and brain region. Tranylcypromine 38-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 9150301-1 1997 This study compares the effects of two non-steroidal anti-inflammatory drugs, Bufferin A (BA) and L-5409709 (L-54), on nociceptive behaviour and spinal Fos expression induced by subcutaneous formalin in the rat. Barium 90-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-155 9150301-1 1997 This study compares the effects of two non-steroidal anti-inflammatory drugs, Bufferin A (BA) and L-5409709 (L-54), on nociceptive behaviour and spinal Fos expression induced by subcutaneous formalin in the rat. Formaldehyde 191-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-155 9106474-0 1997 Lactation reduces Fos induction in the paraventricular and supraoptic nuclei of the hypothalamus after urethane administration in rats. Urethane 103-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 9106474-2 1997 In these studies we determined whether Fos induction following treatment with urethane would differ between nonlactating and lactating rats. Urethane 78-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 9106474-3 1997 Urethane treatment produced robust Fos expression in the central nucleus of the amygdala and the paraventricular and supraoptic nuclei of the hypothalamus 1 h after treatment. Urethane 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 9106474-5 1997 Removing litters from lactating rats 48 h prior to urethane administration restored the Fos response in the parvocellular division of the paraventricular nucleus (PVN) to the level seen in nonlactating rats. Urethane 51-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 9131661-6 1997 However, when blood glucose was allowed to decrease, Fos expression did increase in the arcuate nucleus and in the brainstem. Glucose 20-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 9098565-0 1997 Induction of preconvulsive behavior and Fos expression by dopamine-induced nigral lesion in the rat. Dopamine 58-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 9098575-1 1997 The effects of inhalation anesthetics, nitrous oxide (N2O) and halothane, on the expression of c-Fos protein evoked by formalin injection were studied in the spinal cord in the rat. Nitrous Oxide 39-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 9098575-1 1997 The effects of inhalation anesthetics, nitrous oxide (N2O) and halothane, on the expression of c-Fos protein evoked by formalin injection were studied in the spinal cord in the rat. Nitrous Oxide 54-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 9098575-1 1997 The effects of inhalation anesthetics, nitrous oxide (N2O) and halothane, on the expression of c-Fos protein evoked by formalin injection were studied in the spinal cord in the rat. Halothane 63-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 9098575-1 1997 The effects of inhalation anesthetics, nitrous oxide (N2O) and halothane, on the expression of c-Fos protein evoked by formalin injection were studied in the spinal cord in the rat. Formaldehyde 119-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 9098575-2 1997 The expression of c-Fos protein was detected by immunocytochemistry following the injection of formalin (5%, 100 microliters) into the plantar surface of the left hindpaw. Formaldehyde 95-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 9098575-8 1997 Both N2O and halothane suppressed the expression of c-Fos in the neck of the dorsal horn and ventral gray in a dose-dependent manner, but no effects were seen at the superficial layer or nucleus proprius. Nitrous Oxide 5-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 9098575-8 1997 Both N2O and halothane suppressed the expression of c-Fos in the neck of the dorsal horn and ventral gray in a dose-dependent manner, but no effects were seen at the superficial layer or nucleus proprius. Halothane 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 9098575-9 1997 Suppression of c-Fos expression was greater under N2O than halothane anesthesia. Nitrous Oxide 50-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 9098575-9 1997 Suppression of c-Fos expression was greater under N2O than halothane anesthesia. Halothane 59-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 9060477-5 1997 Neuroprotective concentrations of CHX caused only a moderate (20-40%) reduction in overall protein synthesis, and induced an increase in c-fos, c-jun, and bcl-2 mRNAs and protein levels as determined by reverse transcription-PCR analysis and immunocytochemistry, respectively. Cycloheximide 34-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 9070740-2 1997 Recent evidence indicates that the paracrine-acting messenger molecule nitric oxide may be involved in the activation or regulation of c-fos expression in neurons, and many neurons in the paraventricular nucleus contain the enzyme nitric oxide synthase. Nitric Oxide 71-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 9070740-4 1997 To study whether nitric oxide is involved in stress activation of c-fos expression in the paraventricular nucleus, we assessed the effect of treatment with competitive nitric oxide synthase blockers on expression of Fos protein in neurons of the paraventricular nucleus of rats subjected to immobilization stress. Nitric Oxide 17-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 9070740-7 1997 These results indicate that nitric oxide is involved in the regulation of Fos expression in stress-activated cells of the paraventricular nucleus. Nitric Oxide 28-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 9070758-0 1997 Morphine and somatostatin analogue reduce c-fos expression in trigeminal subnucleus caudalis produced by corneal stimulation in the rat. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 9070758-4 1997 Morphine reduced the number of Fos-positive neurons produced at the transition region between trigeminal subnucleus caudalis and the upper cervical spinal cord, whereas c-fos expression at the subnucleus interpolaris/caudalis transition was not affected significantly. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 9070758-5 1997 Morphine also reduced the arterial pressure and heart rate responses to corneal stimulation in proportion to the dose of morphine and required a threshold dose similar to that which reduced c-fos expression. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 190-195 9070758-6 1997 Naloxone prevented the morphine-induced inhibition of c-fos expression and cardiovascular reflex responses to corneal stimulation. Naloxone 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 9098543-0 1997 Cryptic brain cell injury caused by fetal nicotine exposure is associated with persistent elevations of c-fos protooncogene expression. Nicotine 42-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 9070758-6 1997 Naloxone prevented the morphine-induced inhibition of c-fos expression and cardiovascular reflex responses to corneal stimulation. Morphine 23-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 9070758-9 1997 A subthreshold dose of morphine plus a threshold dose of somatostatin analogue caused a greater inhibition of Fos-positive neurons at the subnucleus caudalis/cervical cord transition, but not in reflex-evoked autonomic responses, than the same dose of either drug alone. Morphine 23-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 9098543-2 1997 In the current study, pregnant rats were given nicotine by implanted minipump infusion either from gestational days 4-12 or 4-21 and fetal and neonatal brain regions were examined for expression of the mRNA encoding c-fos, a nuclear transcription factor that becomes chronically elevated when cell injury or apoptosis are occurring. Nicotine 47-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 216-221 9098543-5 1997 In contrast to the elevation of c-fos seen with prenatal nicotine, postnatal nicotine injections given to 2-day-old rats did not cause acute stimulation of c-fos expression. Nicotine 57-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 9098543-6 1997 The ability of injected nicotine to evoke acute rises in c-fos emerged by postnatal day 8 and initially displayed regional specificity paralleling the concentration of nicotinic cholinergic receptors. Nicotine 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 9098543-7 1997 With increasing maturity, regional selectivity of the c-fos response to acute nicotine was lost, consistent with indirect actions that could be mediated through nicotine-induced hypoxia/ischemia. Nicotine 78-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 9098523-0 1997 Effects of neuroprotective dose of fructose-1,6-bisphosphate on hypoxia-induced expression of c-fos and hsp70 mRNA in neonatal rat cerebrocortical slices. fructose-1,6-diphosphate 35-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 9098543-8 1997 These results indicate that prenatal nicotine exposure causes chronic elevations of c-fos expression in fetal and neonatal brain that are distinguishable from the later onset of the ability of acute nicotine to cause short-term stimulation of c-fos. Nicotine 37-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 9098543-8 1997 These results indicate that prenatal nicotine exposure causes chronic elevations of c-fos expression in fetal and neonatal brain that are distinguishable from the later onset of the ability of acute nicotine to cause short-term stimulation of c-fos. Nicotine 37-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 243-248 9098543-8 1997 These results indicate that prenatal nicotine exposure causes chronic elevations of c-fos expression in fetal and neonatal brain that are distinguishable from the later onset of the ability of acute nicotine to cause short-term stimulation of c-fos. Nicotine 199-207 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 243-248 9098543-10 1997 Given that chronic elevations of c-fos are known to be associated with cell injury and to evoke apoptosis in otherwise healthy cells, these results suggest that prenatal nicotine exposure evokes delayed neurotoxicity by altering the program of neural cell differentiation, and that elevated c-fos expression provides an early marker of the eventual deficits. Nicotine 170-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 9098543-10 1997 Given that chronic elevations of c-fos are known to be associated with cell injury and to evoke apoptosis in otherwise healthy cells, these results suggest that prenatal nicotine exposure evokes delayed neurotoxicity by altering the program of neural cell differentiation, and that elevated c-fos expression provides an early marker of the eventual deficits. Nicotine 170-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 291-296 9098550-1 1997 We have previously shown that systemic administration of non-selective dopamine agonists results in a pronounced expression of the proto-oncoprotein Fos within the lateral habenula. Dopamine 71-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 149-152 9132683-0 1997 A blocker of nitric oxide synthase, NG-nitro-L-arginine methyl ester, attenuates light-induced Fos protein expression in rat suprachiasmatic nucleus. NG-Nitroarginine Methyl Ester 36-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 9089206-0 1997 Inhibition of carrageenan-induced spinal c-Fos activation by systemically administered c-fos antisense oligodeoxynucleotides may be facilitated by local opening of the blood-spinal cord barrier. Carrageenan 14-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 9089206-0 1997 Inhibition of carrageenan-induced spinal c-Fos activation by systemically administered c-fos antisense oligodeoxynucleotides may be facilitated by local opening of the blood-spinal cord barrier. Carrageenan 14-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 9089206-0 1997 Inhibition of carrageenan-induced spinal c-Fos activation by systemically administered c-fos antisense oligodeoxynucleotides may be facilitated by local opening of the blood-spinal cord barrier. Oligodeoxyribonucleotides 103-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 9089206-0 1997 Inhibition of carrageenan-induced spinal c-Fos activation by systemically administered c-fos antisense oligodeoxynucleotides may be facilitated by local opening of the blood-spinal cord barrier. Oligodeoxyribonucleotides 103-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 9089206-4 1997 In the present study the effect of systemically administered c-fos antisense oligodeoxynucleotides (ODNs) on c-Fos and dynorphin protein levels in rat L4 spinal cord has been investigated by immunohistochemistry during carrageenan-induced hindpaw inflammation. Oligodeoxyribonucleotides 77-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 9089206-4 1997 In the present study the effect of systemically administered c-fos antisense oligodeoxynucleotides (ODNs) on c-Fos and dynorphin protein levels in rat L4 spinal cord has been investigated by immunohistochemistry during carrageenan-induced hindpaw inflammation. Carrageenan 219-230 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 9132683-3 1997 In the present study we investigated this hypothesis by assessing in rats the effect of treatment with the NOS blocker, NG-nitro-L-arginine methyl ester (L-NAME), on light-induced expression of the transcription factor Fos, a cellular marker of light signaling in the SCN. NG-Nitroarginine Methyl Ester 120-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 219-222 9132683-3 1997 In the present study we investigated this hypothesis by assessing in rats the effect of treatment with the NOS blocker, NG-nitro-L-arginine methyl ester (L-NAME), on light-induced expression of the transcription factor Fos, a cellular marker of light signaling in the SCN. NG-Nitroarginine Methyl Ester 154-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 219-222 9132683-4 1997 We found that systemic administration of L-NAME (100 mg/kg) but not of the inactive analog, D-NAME, significantly attenuates light-induced expression of Fos immunoreactivity in the SCN. NG-Nitroarginine Methyl Ester 41-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-156 9030636-0 1997 Phosphorylation of transcription factor CREB in rat spinal cord after formalin-induced hyperalgesia: relationship to c-fos induction. Formaldehyde 70-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 9030636-8 1997 In contrast to CREB phosphorylation, the induction of c-Fos expression reached peak levels 2 hr after formalin treatment and c-Fos induction was mainly ipsilateral. Formaldehyde 102-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 9030636-9 1997 Both formalin-evoked CREB phosphorylation and c-Fos expression in the spinal cord were suppressed by pretreatment with the NMDA receptor antagonist MK-801 (3.5 mg/kg, i.p.) Dizocilpine Maleate 148-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 9058375-4 1997 CAI treatment specifically inhibited thapsigargin- or endothelin-stimulated expression from the c-fos promoter in Rat-1 cells and in epithelial cell lines derived from ovary and breast. carboxyamido-triazole 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 9058375-4 1997 CAI treatment specifically inhibited thapsigargin- or endothelin-stimulated expression from the c-fos promoter in Rat-1 cells and in epithelial cell lines derived from ovary and breast. Thapsigargin 37-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 9003014-0 1997 Induction of c-fos messenger ribonucleic acid in neuropeptide Y and growth hormone (GH)-releasing factor neurons in the rat arcuate nucleus following systemic injection of the GH secretagogue, GH-releasing peptide-6. ribonucleic 29-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 9087636-6 1997 Finally, intragastric hypertonic saline significantly increased Fos immunoreactivity in the nucleus of the solitary tract (NTS), area postrema (AP), lateral parabrachial nucleus (LPBN), supraoptic nucleus (SON), and paraventricular nucleus (PVN). Sodium Chloride 33-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 9175622-2 1997 These results indicate that c-fos induction after ECS may be mediated by Ser-133 phosphorylation of CREB in rat hippocampus, but not in the cerebellum. Serine 73-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 9472398-2 1997 Fos immunoreactivity was mapped in forebrain in unrestrained rats, previously prepared with an indwelling venous catheter, after the intravenous administration of kainic acid (10 mg/kg). Kainic Acid 163-174 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9138721-0 1997 Modulatory role of catecholamines in the transsynaptic expression of c-fos in the rat medial prefrontal cortex induced by disinhibition of the mediodorsal thalamus: a study employing microdialysis and immunohistochemistry. Catecholamines 19-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 9138721-3 1997 Bicuculline perfusion induced Fos-like immunoreactivity in the thalamic projection areas, including the PFC, and in the thalamic nuclei surrounding the dialysis probe. Bicuculline 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 9138721-5 1997 However, densitometric image analysis revealed that the intensity of Fos-like immunoreactivity in the PFC of lesioned rats perfused with 0.1 mM bicuculline was higher than in correspondingly treated controls. Bicuculline 144-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 9138721-8 1997 The increase in the intensity of Fos-like immunostaining in strongly stimulated, catecholamine-depleted rats suggests that catecholamines modulate the degree to which thalamic activity can activate the PFC of awake animals. Catecholamines 81-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 9138721-8 1997 The increase in the intensity of Fos-like immunostaining in strongly stimulated, catecholamine-depleted rats suggests that catecholamines modulate the degree to which thalamic activity can activate the PFC of awake animals. Catecholamines 123-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 9034904-5 1997 Thus, we reasoned that the earliest brain regions to express Fos might be involved in the initiation and maintenance of soman-induced convulsions. Soman 120-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 9034904-14 1997 The rapid induction of Fos in the piriform cortex and the locus coeruleus, taken together with previous anatomical, eletrophysiological and neurochemical studies, suggests that prolonged, excessive exposure to synaptically released acetylcholine and norepinephrine triggers the production of soman-induced seizures initially in the piriform cortex and subsequently in other cortical and subcortical structures. Acetylcholine 232-245 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 9034904-14 1997 The rapid induction of Fos in the piriform cortex and the locus coeruleus, taken together with previous anatomical, eletrophysiological and neurochemical studies, suggests that prolonged, excessive exposure to synaptically released acetylcholine and norepinephrine triggers the production of soman-induced seizures initially in the piriform cortex and subsequently in other cortical and subcortical structures. Norepinephrine 250-264 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 9034904-14 1997 The rapid induction of Fos in the piriform cortex and the locus coeruleus, taken together with previous anatomical, eletrophysiological and neurochemical studies, suggests that prolonged, excessive exposure to synaptically released acetylcholine and norepinephrine triggers the production of soman-induced seizures initially in the piriform cortex and subsequently in other cortical and subcortical structures. Soman 292-297 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 9063681-0 1997 Adenosine A2A receptor antagonism potentiates L-DOPA-induced turning behaviour and c-fos expression in 6-hydroxydopamine-lesioned rats. Oxidopamine 103-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 9063681-5 1997 Expression of c-fos as measured by Fos-like immunoreactivity after SCH 58261 plus L-DOPA was also potentiated as compared with L-DOPA alone, both in striatum and globus pallidus of the 6-hydroxydopamine-lesioned side of the brain. Levodopa 82-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 9063681-5 1997 Expression of c-fos as measured by Fos-like immunoreactivity after SCH 58261 plus L-DOPA was also potentiated as compared with L-DOPA alone, both in striatum and globus pallidus of the 6-hydroxydopamine-lesioned side of the brain. Levodopa 127-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 9063681-5 1997 Expression of c-fos as measured by Fos-like immunoreactivity after SCH 58261 plus L-DOPA was also potentiated as compared with L-DOPA alone, both in striatum and globus pallidus of the 6-hydroxydopamine-lesioned side of the brain. Oxidopamine 185-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 9070635-0 1997 Enhanced CREB phosphorylation and changes in c-Fos and FRA expression in striatum accompany amphetamine sensitization. Amphetamine 92-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 9070635-3 1997 Similar to previous observations using chronic cocaine administration, amphetamine sensitized animals had decreased c-Fos and increased FRA proteins in striatum. Amphetamine 71-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 9067440-0 1997 Corticotropin-releasing factor and systemic capsaicin-sensitive afferents are involved in abdominal surgery-induced Fos expression in the paraventricular nucleus of the hypothalamus. Capsaicin 44-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 9067440-7 1997 Capsaicin (125 mg/kg s.c., 2 weeks before) or alpha-helical CRF9-41 (50 microg i.c.v., before surgery) reduced the number of Fos-positive cells by 50% in the PVN while not modifying the number of Fos-labelled cells in the other nuclei. Capsaicin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-128 9067440-7 1997 Capsaicin (125 mg/kg s.c., 2 weeks before) or alpha-helical CRF9-41 (50 microg i.c.v., before surgery) reduced the number of Fos-positive cells by 50% in the PVN while not modifying the number of Fos-labelled cells in the other nuclei. Capsaicin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 196-199 9067450-0 1997 Intracarotid glucose selectively increases Fos-like immunoreactivity in paraventricular, ventromedial and dorsomedial nuclei neurons. Glucose 13-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 9045998-7 1997 The effects of intravenous phenoxybenzamine were different, in that at all dose levels phenoxybenzamine completely blocked the LH surge and reduced by approximately half, the cFos activation in LHRH neurons (independent of dose). Phenoxybenzamine 87-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 175-179 9106764-0 1997 Bicuculline infusion advances the timing of Fos expression in LHRH neurons in the preoptic area of proestrous rats. Bicuculline 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 9106764-1 1997 The effect of bicuculline (BIC) on Fos expression in lutenizing hormone-releasing hormone (LHRH) neurons was examined by immunocytochemistry. Bicuculline 14-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 9106764-1 1997 The effect of bicuculline (BIC) on Fos expression in lutenizing hormone-releasing hormone (LHRH) neurons was examined by immunocytochemistry. Bicuculline 27-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 9106764-3 1997 The distribution and proportion of LHRH neurons expressing Fos in the BIC 1400 group were identical to those in the SAL 1700 group. Bicuculline 70-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 9058289-4 1997 In an injured state following indomethacin treatment, reg gene expression was markedly augmented, accompanied by an increase of c-fos expression and healing of the mucosal lesions. Indomethacin 30-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-133 9039826-8 1997 Genistein (750 microg/g) induced the uterine expression of c-fos. Genistein 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 9041360-0 1997 Fos expression by naloxone in LHRH neurons of the mediobasal hypothalamus and effects of pentobarbital sodium in the proestrous rat. Naloxone 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9041360-2 1997 Further, we examined whether an ovulation-blocking dosage of pentobarbital sodium (PB) would affect the NAL-induced Fos expression. Pentobarbital 61-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 9041360-2 1997 Further, we examined whether an ovulation-blocking dosage of pentobarbital sodium (PB) would affect the NAL-induced Fos expression. Pentobarbital 83-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 9041360-2 1997 Further, we examined whether an ovulation-blocking dosage of pentobarbital sodium (PB) would affect the NAL-induced Fos expression. Naloxone 104-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 9041360-8 1997 15 min prior to the beginning of NAL infusion significantly enhanced the increase in LH secretion due to NAL, and also enhanced Fos-ir expression in LHRH-ir neurons. Naloxone 33-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-131 9041367-0 1997 Melatonin inhibits GnRH-induced increase of cFOS immunoreactivity in neonatal rat pituitary. Melatonin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-48 9041367-3 1997 The purpose of this study was to determine, whether melatonin inhibits the GnRH-induced induction of cFos in neonatal rat pituitary cells. Melatonin 52-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-105 9041367-4 1997 The effects of GnRH and/or melatonin on cFos immunoreactivity was determined in primary cultures of neonatal rat pituitary cells attached to the coverslip. Melatonin 27-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-44 9041367-9 1997 Melatonin had no effect on basal cFos immunoreactivity, but it inhibited the GnRH-induced (10 nM) increase of cFos in dose-dependent manner (EC50=12 pM). Melatonin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-114 9027733-12 1997 We also found that the genes that encode the c-Fos and JunB transcription factor proteins are regulated by reduced O2 tension. Oxygen 115-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 9027868-0 1997 Reduced eticlopride-induced Fos expression in caudate-putamen and globus pallidus after unilateral frontal cortex injury: relation to neglect. eticlopride 8-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 9093533-0 1997 cAMP-mediated c-fos expression in pressure-overloaded acceleration of protein synthesis in adult rat heart. Cyclic AMP 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 9093533-1 1997 OBJECTIVES: The aim was to determine whether proto-oncogene c-fos expression and acceleration of protein synthesis by acute pressure overload to the heart were coupled with a cAMP- and protein-kinase-A-dependent system in adult rat heart. Cyclic AMP 175-179 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-65 9093533-12 1997 CONCLUSIONS: These results suggest that c-fos expression induced by acute pressure overload may be coupled with increased cAMP content and protein kinase A activity in addition to increased protein kinase C activity in adult rat heart. Cyclic AMP 122-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 9001250-4 1997 v-Fos-transformed cell invasion is inhibited by c-jun antisense oligonucleotides and by expression of a c-jun dominant negative mutant, TAM-67. Oligonucleotides 64-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 2-5 9001250-8 1997 CD44 antisense oligonucleotides reduce expression of CD44 in v-Fos- or EGF-transformed cells and inhibit invasion but not migration. Oligonucleotides 15-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 9027868-1 1997 Unilateral ablation of medial agranular cortex in rats results in neglect of contralateral stimuli and reductions in amphetamine-induced expression of the immediate early gene, c-fos, in both caudate-putamen and globus pallidus. Amphetamine 117-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 177-182 9027868-2 1997 Both unilateral neglect and the reductions in dopamine agonist induction of subcortical Fos immunoreactivity dissipate over a matter of weeks. Dopamine 46-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 9027868-3 1997 Dopamine agonism induces Fos predominantly in striatonigral cells and in globus pallidus via striatopallidal disinhibition, whereas Fos is induced in striatopallidal cells by administration of antagonists of the D2 dopamine receptor subfamily. Dopamine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 9027868-4 1997 To examine more directly effects of cortical injury on striatopallidal function, induction of striatal Fos by the D2 antagonist eticlopride (0.1 mg/kg, s.c.) was examined in rats with medial agranular cortex ablation. eticlopride 128-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-106 9027868-5 1997 In the same animals, eticlopride-induced Fos in globus pallidus was also examined. eticlopride 21-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 9027868-6 1997 Five days after unilateral cortex injury, in rats showing neglect, the numbers of Fos immunoreactive nuclei induced by eticlopride were reduced by 50% in caudate-putamen and 25% in globus pallidus of the ipsilateral hemisphere. eticlopride 119-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 9027868-8 1997 Three or more weeks after cortical injury, in rats recovered from neglect, eticlopride-induced Fos was normalized in caudate-putamen, but still decreased by 20% in globus pallidus. eticlopride 75-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 9027869-0 1997 Amphetamine sensitization enhances regional c-fos expression produced by conditioned fear. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 9027869-2 1997 In an attempt to investigate the neurobiological correlates of this phenomenon, the present study examined the effects of prior D-amphetamine sensitization on regional c-fos expression induced by a psychological stressor. Dextroamphetamine 128-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-173 9027869-7 1997 The amphetamine sensitization procedure significantly enhanced the effects of conditioned fear on c-fos expression in several brain regions. Amphetamine 4-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 9135060-3 1997 The distribution of alpha1B receptor immunoreactivity overlapped in part with the distributions of c-Fos immunoreactivity induced in the paraventricular nucleus by either restraint stress or hypertonic saline administration. Sodium Chloride 202-208 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 9042522-4 1997 However, dexamethasone did inhibit the induction of c-fos transcription seen following HIE, and subsequent evidence of apoptosis. Dexamethasone 9-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 9042526-0 1997 Distribution of Fos-immunoreactivity in rat brain following a dipsogenic dose of captopril and effects of angiotensin receptor blockade. Captopril 81-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 9042526-1 1997 Immunohistochemical techniques were used to detect Fos in the brain following subcutaneous administration of the angiotensin converting enzyme inhibitors captopril or enalapril at 0.5 mg/kg to conscious rats. Captopril 154-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 9042526-4 1997 Pre-treatment with the angiotensin AT1 receptor antagonist ZD7155 (10 mg/kg) given subcutaneously prevented the captopril-induced increase in Fos in the lamina terminalis. ZD 7155 59-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-145 9042526-4 1997 Pre-treatment with the angiotensin AT1 receptor antagonist ZD7155 (10 mg/kg) given subcutaneously prevented the captopril-induced increase in Fos in the lamina terminalis. Captopril 112-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-145 9042531-1 1997 After acquisition of a conditioned taste aversion (CTA) against sucrose, intraoral infusions of sucrose induce c-Fos-like immunoreactivity (c-FLI) in the medial intermediate nucleus of the solitary tract (iNTS) of the rat. Sucrose 96-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 9037495-3 1997 Treatment with a behaviorally-effective dose of estradiol increases Fos expression, suggestive of neuronal response, and subsequent treatment with a behaviorally-effective dose of progesterone further increases Fos expression within a few hours in female rat brain. Estradiol 48-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 9040699-14 1997 The signaling mechanisms underlying the effect of fasting and subsequent lowering of plasma glucose levels on postischemic c-fos expression remain to be explored. Glucose 92-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 9037495-3 1997 Treatment with a behaviorally-effective dose of estradiol increases Fos expression, suggestive of neuronal response, and subsequent treatment with a behaviorally-effective dose of progesterone further increases Fos expression within a few hours in female rat brain. Estradiol 48-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 211-214 9037495-4 1997 In order to determine if neurons that respond to progesterone with increase Fos expression also contain progestin receptors, we used a double-label immunofluorescent technique to label both progestin receptors and Fos protein following progesterone or vehicle treatment of estradiol-primed female rats. Progesterone 49-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 9037495-5 1997 As shown previously, progesterone treatment increased Fos expression in progestin receptor-containing regions, such as the ventromedial nucleus of the hypothalamus and the medial preoptic area. Progesterone 21-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 9037495-6 1997 In addition, progesterone treatment induced a statistically-significant increase in Fos-immunoreactivity within progestin receptor-containing cells in the medial preoptic area and the ventromedial nucleus of the hypothalamus, but not in the arcuate nucleus. Progesterone 13-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 9037495-8 1997 The progesterone-induced Fos expression within progestin receptor-containing neurons may or may not be associated with the effects of progesterone on sexual or other reproductive behaviors, as it remains to be tested. Progesterone 4-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 9037495-9 1997 However, the Fos expression provides a useful marker to aid in identification of neurons that respond to a behaviorally-relevant dose of progesterone. Progesterone 137-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 9037396-1 1997 In this study, we monitored Fos-like immunoreactivity in the sacral spinal cord to identify neurons that are likely to contribute to the autonomic manifestations of opioid antagonist-precipitated withdrawal in morphine-tolerant rats. Morphine 210-218 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 9038892-4 1997 In in vitro studies, exposure of control tissue to 50 mM K(+)-Krebs-EA (2 x 10(-5) M) solutions significantly increased Fos immunoreactivity in the myenteric plexus, whereas a basal level of Fos was seen in control tissue incubated in Krebs solution, sham-sensitized tissue exposed to bovine serum albumin (BSA, 2 x 10(-5) M), or EA and sensitized tissue exposed to BSA. k(+)-krebs 57-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-123 9121675-0 1997 A contribution of neurokinin-1 receptor to formalin-induced c-fos expression in the rat spinal dorsal horn. Formaldehyde 43-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 9121675-1 1997 Formalin applied to a hindpaw of the rat resulted in c-fos expression in the spinal dorsal horn neurons on the ipsilateral side of stimulation. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 9121675-3 1997 L-668169 administered intrathecally (1 microg/10 microl, 10 microg/10 microl) 15 min before injection of 2% formalin into one hindpaw significantly decreased the number of Fos-labeled neurons in the dorsal horn. L 668169 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-175 9121675-5 1997 It was also showed that a selective NK-1 agonist Sar-SP perfused intrathecally (10 microg/10 microl) did result in the production of c-fos mainly in laminae I and that SP-like immunoreactive varicosities apposed neurons with Fos-labeled nuclei at their perikaryal membrane in the superficial laminae. sar-sp 49-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 9121675-5 1997 It was also showed that a selective NK-1 agonist Sar-SP perfused intrathecally (10 microg/10 microl) did result in the production of c-fos mainly in laminae I and that SP-like immunoreactive varicosities apposed neurons with Fos-labeled nuclei at their perikaryal membrane in the superficial laminae. sar-sp 49-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 225-228 9121675-5 1997 It was also showed that a selective NK-1 agonist Sar-SP perfused intrathecally (10 microg/10 microl) did result in the production of c-fos mainly in laminae I and that SP-like immunoreactive varicosities apposed neurons with Fos-labeled nuclei at their perikaryal membrane in the superficial laminae. TFF2 protein, human 53-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 9121675-5 1997 It was also showed that a selective NK-1 agonist Sar-SP perfused intrathecally (10 microg/10 microl) did result in the production of c-fos mainly in laminae I and that SP-like immunoreactive varicosities apposed neurons with Fos-labeled nuclei at their perikaryal membrane in the superficial laminae. TFF2 protein, human 53-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 225-228 8974398-1 1997 Regional brain activation was assessed by mapping of Fos-related protein expression in rats trained to self-administration of intravenous nicotine and cocaine. Nicotine 138-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 8974398-1 1997 Regional brain activation was assessed by mapping of Fos-related protein expression in rats trained to self-administration of intravenous nicotine and cocaine. Cocaine 151-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 9042442-0 1997 Electrically evoked auditory brainstem response and Fos-immunoreactivity in kanamycin-deafened rats. Kanamycin 76-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 9039019-5 1997 Magnocellular neurons expressing Fos in the SON increased linearly with plasma osmolality and were more numerous (P < 0.05) in control lactating animals but increased less (P < 0.05) than in virgin rats after 0.85 M NaCl. Sodium Chloride 222-226 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 9227847-9 1997 The level of c-fos mRNA was increased only the dorsal striatum following morphine injections. Morphine 73-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 9067632-3 1997 Similar to IgE-mediated cell degranulation we demonstrated inhibition of the c-fos signal in the absence of calcium and after preincubation of cells with the protein tyrosine kinase inhibitor genistein. Genistein 192-201 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 9067632-5 1997 Depletion of protein kinase C by PMA pre-treatment resulted in substantial inhibition of the c-fos signal. Tetradecanoylphorbol Acetate 33-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 9067632-7 1997 In addition, we demonstrate that c-fos mRNA expression is not restricted to Fc epsilon RI activation but can be induced by a variety of IgE independent mechanisms including calcium influx by ionophore A 23187 and stimulation of G proteins. Calcium 173-180 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 9042566-0 1997 Prevention of kainic acid-induced limbic seizures and Fos expression by the GABA-A receptor agonist muscimol. Muscimol 100-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 9042566-5 1997 Following 0.5 mg/kg muscimol treatment a remarkable decrease of Fos expression occurred but only in the caudate putamen and core of the accumbens nucleus. Muscimol 20-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 9042566-6 1997 Treatment with 1 mg/kg muscimol led to further significant decreases of Fos expression in CA1-3 pyramidal neurons and the disappearance of Fos induction in the cerebral cortex above the rhinal fissure, reticular thalamic nucleus, claustrum, fundus striati, ventral pallidum, septal nucleus, lateral habenular nucleus, and lateral amygdaloid nucleus. Muscimol 23-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 9042566-6 1997 Treatment with 1 mg/kg muscimol led to further significant decreases of Fos expression in CA1-3 pyramidal neurons and the disappearance of Fos induction in the cerebral cortex above the rhinal fissure, reticular thalamic nucleus, claustrum, fundus striati, ventral pallidum, septal nucleus, lateral habenular nucleus, and lateral amygdaloid nucleus. Muscimol 23-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 9042566-7 1997 When 2 mg/kg muscimol was injected, animals exhibited "absence seizures" instead of limbic seizures, and Fos expression in the hippocampus was effectively blocked. Muscimol 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-108 9042572-0 1997 5-Hydroxytryptamine induces TIS8/egr-1 and c-fos expression in PC12 cells. Serotonin 0-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 15102416-3 1997 Lesioning the VLMIat in hypertensive rats by unilateral quinolinic acid (QA) injection (0.3 microl of a 180 nmol/microl solution) 24 h before noxious stimulation, prevented the Fos-IR cell decrease. Quinolinic Acid 56-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 177-180 15102416-3 1997 Lesioning the VLMIat in hypertensive rats by unilateral quinolinic acid (QA) injection (0.3 microl of a 180 nmol/microl solution) 24 h before noxious stimulation, prevented the Fos-IR cell decrease. Quinolinic Acid 73-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 177-180 9403355-2 1997 The aim of this study was to verify if analgesic drugs, such as morphine and ketorolac, may affect the c-fos protooncogene expression by using a method highly sensitive and specific, based on transformation of activated c-fos specific mRNA in cDNA (reverse transcription), its amplification (PCR) and final visualization by electrophoresis on agarose gel. Morphine 64-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 9403355-2 1997 The aim of this study was to verify if analgesic drugs, such as morphine and ketorolac, may affect the c-fos protooncogene expression by using a method highly sensitive and specific, based on transformation of activated c-fos specific mRNA in cDNA (reverse transcription), its amplification (PCR) and final visualization by electrophoresis on agarose gel. Morphine 64-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 220-225 9403355-2 1997 The aim of this study was to verify if analgesic drugs, such as morphine and ketorolac, may affect the c-fos protooncogene expression by using a method highly sensitive and specific, based on transformation of activated c-fos specific mRNA in cDNA (reverse transcription), its amplification (PCR) and final visualization by electrophoresis on agarose gel. Ketorolac 77-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 9403355-2 1997 The aim of this study was to verify if analgesic drugs, such as morphine and ketorolac, may affect the c-fos protooncogene expression by using a method highly sensitive and specific, based on transformation of activated c-fos specific mRNA in cDNA (reverse transcription), its amplification (PCR) and final visualization by electrophoresis on agarose gel. Ketorolac 77-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 220-225 9403355-2 1997 The aim of this study was to verify if analgesic drugs, such as morphine and ketorolac, may affect the c-fos protooncogene expression by using a method highly sensitive and specific, based on transformation of activated c-fos specific mRNA in cDNA (reverse transcription), its amplification (PCR) and final visualization by electrophoresis on agarose gel. Sepharose 343-350 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 9403355-2 1997 The aim of this study was to verify if analgesic drugs, such as morphine and ketorolac, may affect the c-fos protooncogene expression by using a method highly sensitive and specific, based on transformation of activated c-fos specific mRNA in cDNA (reverse transcription), its amplification (PCR) and final visualization by electrophoresis on agarose gel. Sepharose 343-350 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 220-225 9403355-4 1997 When the animals were pretreated with morphine or ketorolac and subsequently exposed to the monolateral sciatic nerve ligature, or treated with ketorolac immediately after the same painful stimulus, we found that only pretreatment with morphine completely blocked c-fos depression. Morphine 236-244 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 264-269 17657612-0 1997 Potent anti-inflammatory/analgesic effects of lornoxicam in comparison to other nsaids: a c-fos study in the rat. lornoxicam 46-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 17657612-1 1997 This study evaluates the anti-inflammatory/analgesic effects of lornoxicam, a new non-steroidal anti-inflammatory drug, using the method of c-Fos protein immunoreactivity in the carrageenan model of inflammatory nociception in the rat. lornoxicam 64-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 17657612-5 1997 Preadministered lornoxicam dose relatedly reduced the total number of c-Fos-LI neurons (regression coefficient r=0.79; p<0.001) with the strongest effect corresponding to the 75+/-2% reduction (p<0.001) for the highest dose of 9 mg/kg, and the 45+/-3% reduction (p<0.001) for the low dose of 0.3 mg/kg. lornoxicam 16-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 17657612-8 1997 The lowes dose of lornoxicam (0.1 mg/kg iv) had a similar effect in both superficial and deep laminae, whereas the four higher doses (0.3, 1, 3 and 9 mg/kg iv) had a significantly stronger effect on the number of c-Fos-LI neurons in deep laminae as compared to that in superficial laminae. lornoxicam 18-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-218 9350507-2 1997 Approximately 32% of the neurons with NKR-LI in the NST expressed c-fos protein after chemical irritation on the peritoneum with acetic acid. Acetic Acid 129-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 10456107-3 1997 To address this question in the present study, we have used a synthetic antisense phosphorothioate oligodeoxynucleotide to suppress in vivo the expression of c-fos in rat brain. phosphorothioate oligodeoxynucleotide 82-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 10456107-4 1997 Intrahippocampal application of the oligodeoxynucleotide 10 hr and 2 hr before starting a brightness discrimination training drastically reduced the induction of c-Fos immunoreactivity normally observed in limbic and cortical areas after the training session. Oligodeoxyribonucleotides 36-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 9154216-2 1997 To identify these targets we have mapped alcohol-induced changes in the expression of the c-Fos protein in the rat brain. Alcohols 41-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 8983033-5 1997 Amphetamine also induced different patterns of Fos immunoreactivity in the neostriatum and nucleus accumbens of young and aged rats, as Fos expression in aged rats exhibited a distinctive dorsal to ventral pattern of decline. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 8983033-7 1997 In contrast, the D2 agonist quinpirole substantially enhanced the motor activity and Fos expression of young rats, while only modestly affecting aged rats. Quinpirole 28-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 9131593-2 1997 Low doses of domoic acid (0.1 mg/kg) induced c-fos in the central nervous system which was inhibited in part by 2-amino-5-phosphonovaleric acid, an NMDA receptor antagonist. domoic acid 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 9131593-2 1997 Low doses of domoic acid (0.1 mg/kg) induced c-fos in the central nervous system which was inhibited in part by 2-amino-5-phosphonovaleric acid, an NMDA receptor antagonist. 2-Amino-5-phosphonovalerate 112-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 9215997-0 1997 Trimethyltin exposure in the rat induces delayed changes in brain-derived neurotrophic factor, fos and heat shock protein 70. trimethyltin 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 9060019-0 1997 Differential contribution of the two phases of the formalin test to the pattern of c-fos expression in the rat spinal cord: studies with remifentanil and lidocaine. Formaldehyde 51-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 9060019-12 1997 In laminae I-II of the L4-L5 segment, the magnitude of the decrease in Fos expression was comparable for remifentanil (26.5%) and lidocaine (27.3%); the decrease was greater when both remifentanil and lidocaine were administered (50.5%), and even greater when bupivicaine and lidocaine were used (74.2%). Remifentanil 105-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 9060019-12 1997 In laminae I-II of the L4-L5 segment, the magnitude of the decrease in Fos expression was comparable for remifentanil (26.5%) and lidocaine (27.3%); the decrease was greater when both remifentanil and lidocaine were administered (50.5%), and even greater when bupivicaine and lidocaine were used (74.2%). Lidocaine 130-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 9060019-12 1997 In laminae I-II of the L4-L5 segment, the magnitude of the decrease in Fos expression was comparable for remifentanil (26.5%) and lidocaine (27.3%); the decrease was greater when both remifentanil and lidocaine were administered (50.5%), and even greater when bupivicaine and lidocaine were used (74.2%). Remifentanil 184-196 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 9060019-12 1997 In laminae I-II of the L4-L5 segment, the magnitude of the decrease in Fos expression was comparable for remifentanil (26.5%) and lidocaine (27.3%); the decrease was greater when both remifentanil and lidocaine were administered (50.5%), and even greater when bupivicaine and lidocaine were used (74.2%). Lidocaine 201-210 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 9060019-12 1997 In laminae I-II of the L4-L5 segment, the magnitude of the decrease in Fos expression was comparable for remifentanil (26.5%) and lidocaine (27.3%); the decrease was greater when both remifentanil and lidocaine were administered (50.5%), and even greater when bupivicaine and lidocaine were used (74.2%). bupivicaine 260-271 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 9060019-12 1997 In laminae I-II of the L4-L5 segment, the magnitude of the decrease in Fos expression was comparable for remifentanil (26.5%) and lidocaine (27.3%); the decrease was greater when both remifentanil and lidocaine were administered (50.5%), and even greater when bupivicaine and lidocaine were used (74.2%). Lidocaine 201-210 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 9060019-13 1997 In laminae V-VI, remifentanil, by itself, decreased c-fos expression by 39.4%; for lidocaine alone, the decrease was 58.4%. Remifentanil 17-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 9016916-1 1996 The effect of acute and chronic administration of dopamine receptor antagonists on the expression of mRNA encoding the cellular immediate-early gene c-fos was investigated in rat brain by in situ hybridization using 35S-labelled oligonucleotide probes. Sulfur-35 216-219 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 149-154 9016916-1 1996 The effect of acute and chronic administration of dopamine receptor antagonists on the expression of mRNA encoding the cellular immediate-early gene c-fos was investigated in rat brain by in situ hybridization using 35S-labelled oligonucleotide probes. Oligonucleotides 229-244 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 149-154 9016916-3 1996 Acute treatment with haloperidol increased the level of c-fos mRNA in the caudate-putamen, nucleus accumbens shell and core, olfactory tubercle and parietal cortex. Haloperidol 21-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 9016916-4 1996 After chronic treatment with haloperidol increases in the level of c-fos mRNA in the caudate-putamen and nucleus accumbens core were no longer observed. Haloperidol 29-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 9016916-6 1996 In the olfactory tubercle, parietal cortex, frontal cortex and cingulate cortex the level of c-fos mRNA was decreased after chronic haloperidol treatment. Haloperidol 132-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 9016916-7 1996 Acute sulpiride treatment reduced the level of c-fos mRNA in the olfactory tubercle, parietal cortex and cingulate cortex. Sulpiride 6-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 9016916-8 1996 After chronic treatment with sulpiride the level of c-fos mRNA was reduced in the dorsal caudate-putamen only. Sulpiride 29-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 9016916-9 1996 Acute clozapine treatment increased the level of c-fos mRNA in the nucleus accumbens shell and islands of Calleja. Clozapine 6-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 9016916-10 1996 After chronic treatment with clozapine the level of c-fos mRNA remained elevated in the islands of Calleja but not in the nucleus accumbens shell. Clozapine 29-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 9016916-12 1996 This study also demonstrated that acute blockade of dopamine receptors with haloperidol, sulpiride and clozapine induced different regionally specific patterns of c-fos expression which were altered after chronic blockade. Haloperidol 76-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-168 9016916-12 1996 This study also demonstrated that acute blockade of dopamine receptors with haloperidol, sulpiride and clozapine induced different regionally specific patterns of c-fos expression which were altered after chronic blockade. Sulpiride 89-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-168 9016916-12 1996 This study also demonstrated that acute blockade of dopamine receptors with haloperidol, sulpiride and clozapine induced different regionally specific patterns of c-fos expression which were altered after chronic blockade. Clozapine 103-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-168 8955147-5 1996 NAC suppressed NGF-induced c-fos gene expression and AP-1 activation. Acetylcysteine 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 8978468-8 1996 Second, the kappa receptor agonist spiradoline (1-10 mg/kg) reduced c-fos and zif 268 induction by SKF-39393 (2.5 mg/kg) preferentially in ventral parts of the dopamine-depleted striatum, which contain higher levels of kappa receptor mRNA and binding. spiradoline 35-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 9051784-3 1996 Supershift analysis with specific antibodies against the members of Fos and Jun protein families showed that the NMDA-induced AP-1 protein complex consisted predominantly of c-Fos and Jun D proteins. N-Methylaspartate 113-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 9051784-3 1996 Supershift analysis with specific antibodies against the members of Fos and Jun protein families showed that the NMDA-induced AP-1 protein complex consisted predominantly of c-Fos and Jun D proteins. N-Methylaspartate 113-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 174-179 21781745-4 1996 Tributyltin-induced c-fos expression was abolished completely by actinomycin D, indicating it was due to transcriptional activation of the gene. Dactinomycin 65-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 9017239-0 1996 Synergistic interaction between an adenosine antagonist and a D1 dopamine agonist on rotational behavior and striatal c-Fos induction in 6-hydroxydopamine-lesioned rats. Adenosine 35-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 9017239-0 1996 Synergistic interaction between an adenosine antagonist and a D1 dopamine agonist on rotational behavior and striatal c-Fos induction in 6-hydroxydopamine-lesioned rats. Oxidopamine 137-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 9017239-3 1996 Coadministration of the adenosine antagonist 3,7-dimethyl-1-propargylxanthine (DMPX: 10 mg/kg) and the D1 dopamine agonist SKF38393 (0.5 mg/kg) to 6-OHDA-lesioned rats produced significant contralateral rotation and c-Fos expression in the ipsilateral striatum compared to 6-OHDA rats treated with either drug alone. 3,7-dimethyl-1-propargylxanthine 45-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 216-221 9017239-4 1996 However, the regional pattern of striatal c-Fos activation following treatment of 6-OHDA rats with SKF38393 and DMPX was different from the dorsolateral pattern of striatal c-Fos induction observed after coadministration of D1 and D2 dopamine agonists (SKF38393: 0.5 mg/kg + quinpirole: 0.05 mg/kg). Oxidopamine 82-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 9017239-4 1996 However, the regional pattern of striatal c-Fos activation following treatment of 6-OHDA rats with SKF38393 and DMPX was different from the dorsolateral pattern of striatal c-Fos induction observed after coadministration of D1 and D2 dopamine agonists (SKF38393: 0.5 mg/kg + quinpirole: 0.05 mg/kg). 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 99-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 9017239-4 1996 However, the regional pattern of striatal c-Fos activation following treatment of 6-OHDA rats with SKF38393 and DMPX was different from the dorsolateral pattern of striatal c-Fos induction observed after coadministration of D1 and D2 dopamine agonists (SKF38393: 0.5 mg/kg + quinpirole: 0.05 mg/kg). 3,7-dimethyl-1-propargylxanthine 112-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 8982717-0 1996 The anti-craving drug acamprosate reduces c-fos expression in rats undergoing ethanol withdrawal. Acamprosate 22-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 8982717-0 1996 The anti-craving drug acamprosate reduces c-fos expression in rats undergoing ethanol withdrawal. Ethanol 78-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 8982717-2 1996 In the present study the expression of the immediate-early gene c-fos in rat hippocampal and cerebellar neurons was used to monitor the modulatory effect of acamprosate on neuronal excitability during ethanol withdrawal. Acamprosate 157-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 8982717-2 1996 In the present study the expression of the immediate-early gene c-fos in rat hippocampal and cerebellar neurons was used to monitor the modulatory effect of acamprosate on neuronal excitability during ethanol withdrawal. Ethanol 201-208 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 8982717-3 1996 Several hybridization techniques were employed to investigate the effect of acamprosate on c-fos expression. Acamprosate 76-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 8982717-4 1996 Acamprosate (200 mg/kg; intraperitoneally) reduced the elevated c-fos mRNA levels in the hippocampus and the cerebellum following 24 h of ethanol withdrawal, or the application of the convulsant pentylenetetrazole. Acamprosate 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 8982717-5 1996 The effect of ethanol withdrawal on c-fos expression was more pronounced in the cerebellum than in the hippocampus. Ethanol 14-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 8982717-6 1996 In the hippocampus (CA1) and the cerebellum acamprosate alone induced a significant increase in c-fos expression in drug-naive animals. Acamprosate 44-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 8982717-7 1996 Only in the hippocampus did co-administration of pentylenetetrazole during ethanol withdrawal induce a further increase in c-fos expression. Pentylenetetrazole 49-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 8982717-7 1996 Only in the hippocampus did co-administration of pentylenetetrazole during ethanol withdrawal induce a further increase in c-fos expression. Ethanol 75-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 8974081-7 1996 While isoproterenol treatment increased steady-state mRNA levels for fos, jun, myc, src, c-erbB-2, ras and topo II, inclusion of pefloxacin with the isoproterenol regimen blocked these increases. Isoproterenol 6-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 9117387-0 1996 Differential effects of haloperidol and two anxiolytic drugs, buspirone and lesopitron, on c-Fos expression in the rat striatum and nucleus accumbens. Haloperidol 24-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 9117387-0 1996 Differential effects of haloperidol and two anxiolytic drugs, buspirone and lesopitron, on c-Fos expression in the rat striatum and nucleus accumbens. Buspirone 62-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 9117387-0 1996 Differential effects of haloperidol and two anxiolytic drugs, buspirone and lesopitron, on c-Fos expression in the rat striatum and nucleus accumbens. lesopitron 76-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 9117387-1 1996 We have studied the effects of the neuroleptic haloperidol and the non-benzodiazepine anxiolytics buspirone and lesopitron on the expression of c-Fos immunoreactivity in the rat forebrain. Haloperidol 47-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-149 9117387-1 1996 We have studied the effects of the neuroleptic haloperidol and the non-benzodiazepine anxiolytics buspirone and lesopitron on the expression of c-Fos immunoreactivity in the rat forebrain. Benzodiazepines 71-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-149 9117387-1 1996 We have studied the effects of the neuroleptic haloperidol and the non-benzodiazepine anxiolytics buspirone and lesopitron on the expression of c-Fos immunoreactivity in the rat forebrain. Buspirone 98-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-149 9117387-1 1996 We have studied the effects of the neuroleptic haloperidol and the non-benzodiazepine anxiolytics buspirone and lesopitron on the expression of c-Fos immunoreactivity in the rat forebrain. lesopitron 112-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-149 9117387-2 1996 Haloperidol and buspirone administration resulted in a significant quantitative increase in the number of Fos-immunoreactive neurons in the lateral striatum and a presumable qualitative increase in the nucleus accumbens. Haloperidol 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 9117387-2 1996 Haloperidol and buspirone administration resulted in a significant quantitative increase in the number of Fos-immunoreactive neurons in the lateral striatum and a presumable qualitative increase in the nucleus accumbens. Buspirone 16-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 9117387-4 1996 The induction of c-Fos immunoreactivity by buspirone is compatible with an interaction of this compound with D2 dopamine receptors, as documented for haloperidol. Buspirone 43-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 9117387-4 1996 The induction of c-Fos immunoreactivity by buspirone is compatible with an interaction of this compound with D2 dopamine receptors, as documented for haloperidol. Haloperidol 150-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 9117393-4 1996 Stress-induced levels of corticosterone were significantly lower in late pregnant and early lactating rats compared with the levels in virgin females, and this correlated with a marked attenuation of stress-induced c-fos mRNA expression in the parvocellular division of the PVN. Corticosterone 25-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 215-220 9117402-1 1996 We assessed the ability of acute peripheral administration of the AT-1 receptor antagonist, losartan, to reverse both the water intake and Fos-immunoreactivity (Fos-ir) induced in rats by either peripheral or cerebroventricular (i.c.v.) Losartan 92-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 9117402-1 1996 We assessed the ability of acute peripheral administration of the AT-1 receptor antagonist, losartan, to reverse both the water intake and Fos-immunoreactivity (Fos-ir) induced in rats by either peripheral or cerebroventricular (i.c.v.) Losartan 92-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-164 9117402-10 1996 Fos-ir was induced by PEG and isoproterenol in several regions of the brain also activated by Ang II. Polyethylene Glycols 22-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9117402-10 1996 Fos-ir was induced by PEG and isoproterenol in several regions of the brain also activated by Ang II. Isoproterenol 30-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9117402-11 1996 Fos-ir was greatly attenuated in the SFO by losartan following administration of PEG, but not isoproterenol, and was either unaffected or increased in SON and PVN after either agent. Losartan 44-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 9117402-11 1996 Fos-ir was greatly attenuated in the SFO by losartan following administration of PEG, but not isoproterenol, and was either unaffected or increased in SON and PVN after either agent. Polyethylene Glycols 81-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 8997275-5 1996 PGF2 alpha also induced the expression of c-fos, atrial natriuretic factor (ANF), and alpha-skeletal actin in these cells. Dinoprost 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 8996800-6 1996 However, striatal Fos activation induced by amphetamine (5 mg/kg i.p., 2 h before killing) revealed that the number of Fos-positive cells detected in the denervated striatal subregion was lower than that observed in the non-denervated one. Amphetamine 44-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 8996800-6 1996 However, striatal Fos activation induced by amphetamine (5 mg/kg i.p., 2 h before killing) revealed that the number of Fos-positive cells detected in the denervated striatal subregion was lower than that observed in the non-denervated one. Amphetamine 44-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-122 8996801-0 1996 Phenotype of striatal cells expressing c-Fos following amphetamine treatment of rats with intrastriatal dopaminergic grafts. Amphetamine 55-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 8996801-1 1996 Activation of the nigrostriatal dopaminergic system by psychostimulants such as amphetamine increases c-Fos expression in the striatum, mostly in the striatonigral substance P-ergic pathway. Amphetamine 80-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 8996801-11 1996 The density of neurons expressing c-Fos following amphetamine treatment was three-fold higher in the graft-bearing striata than in the striata of control animals. Amphetamine 50-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 8996801-13 1996 Similar proportions were found in the graft-bearing striatum, signifying that the pattern of activation of c-fos following amphetamine administration is not changed by the graft. Amphetamine 123-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 8938747-6 1996 The intraperitoneal administration of SR48692, a non-peptide neurotensin receptor antagonist, blocked the neurotensin-induced corticosterone secretion and significantly reduced the number of neurotensin-induced Fos-positive and Zif268-positive neurons in the amygdaloid complex. SR 48692 38-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 211-214 8938747-7 1996 A significant positive correlation was found between the number of Fos-positive nuclei in the central or basolateral nucleus of the amygdala and the serum corticosterone concentration. Corticosterone 155-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 8938747-9 1996 Our findings indicate that the central role of neurotensin in increasing serum corticosterone involves the induction of Fos in the central and basolateral nuclei of the amygdala. Corticosterone 79-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-123 8946502-0 1996 c-Fos induction in the rat nucleus of the solitary tract by intraoral quinine infusion depends on prior contingent pairing of quinine and lithium chloride. Quinine 70-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8946502-0 1996 c-Fos induction in the rat nucleus of the solitary tract by intraoral quinine infusion depends on prior contingent pairing of quinine and lithium chloride. Quinine 126-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8946502-0 1996 c-Fos induction in the rat nucleus of the solitary tract by intraoral quinine infusion depends on prior contingent pairing of quinine and lithium chloride. Lithium Chloride 138-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8946502-1 1996 Intraoral infusions of sucrose or saccharin induce c-Fos-like immunoreactivity (c-FLI) in the intermediate nucleus of the solitary tract (iNTS) of rats after acquisition of a conditioned taste aversion (CTA). Sucrose 23-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 8946502-1 1996 Intraoral infusions of sucrose or saccharin induce c-Fos-like immunoreactivity (c-FLI) in the intermediate nucleus of the solitary tract (iNTS) of rats after acquisition of a conditioned taste aversion (CTA). Saccharin 34-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 8943073-0 1996 Intrastriatally injected c-fos antisense oligonucleotide interferes with striatonigral but not striatopallidal gamma-aminobutyric acid transmission in the conscious rat. Oligonucleotides 41-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 8943073-0 1996 Intrastriatally injected c-fos antisense oligonucleotide interferes with striatonigral but not striatopallidal gamma-aminobutyric acid transmission in the conscious rat. gamma-Aminobutyric Acid 111-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 8943073-1 1996 Antisense c-fos oligonucleotides injected into the neostriatum of conscious rats selectively inhibited c-fos expression associated with compensatory increases in striatal c-fos mRNA levels and also with increased expression of junB and NGFI-A mRNA, probably as a result of regulatory phenomena. Oligonucleotides 16-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-15 8943073-1 1996 Antisense c-fos oligonucleotides injected into the neostriatum of conscious rats selectively inhibited c-fos expression associated with compensatory increases in striatal c-fos mRNA levels and also with increased expression of junB and NGFI-A mRNA, probably as a result of regulatory phenomena. Oligonucleotides 16-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 8943073-1 1996 Antisense c-fos oligonucleotides injected into the neostriatum of conscious rats selectively inhibited c-fos expression associated with compensatory increases in striatal c-fos mRNA levels and also with increased expression of junB and NGFI-A mRNA, probably as a result of regulatory phenomena. Oligonucleotides 16-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 8943073-3 1996 Intrastriatal infusion of the c-fos antisense oligonucleotide profoundly decreased dialysate GABA levels in the ipsilateral substantia nigra within 60 min but did not influence the dialysate GABA levels in the globus pallidus compared with the sham and control oligonucleotide treated groups. Oligonucleotides 46-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 8943073-3 1996 Intrastriatal infusion of the c-fos antisense oligonucleotide profoundly decreased dialysate GABA levels in the ipsilateral substantia nigra within 60 min but did not influence the dialysate GABA levels in the globus pallidus compared with the sham and control oligonucleotide treated groups. gamma-Aminobutyric Acid 93-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 8943073-3 1996 Intrastriatal infusion of the c-fos antisense oligonucleotide profoundly decreased dialysate GABA levels in the ipsilateral substantia nigra within 60 min but did not influence the dialysate GABA levels in the globus pallidus compared with the sham and control oligonucleotide treated groups. Oligonucleotides 261-276 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 8943073-5 1996 The findings demonstrate a facilitatory role for c-fos mediated gene regulation in striatonigral GABA transmission and strengthen the evidence that the regulation of neurotransmission is different in the striatonigral and striatopallidal GABA pathways. gamma-Aminobutyric Acid 97-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 9027330-4 1996 c-fos mRNA levels were decreased 1 h after testosterone treatment in the ventral prostate, whereas they were increased in cycloheximide alone or cycloheximide-testosterone treated groups as compared to vehicle control. Testosterone 43-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9027330-4 1996 c-fos mRNA levels were decreased 1 h after testosterone treatment in the ventral prostate, whereas they were increased in cycloheximide alone or cycloheximide-testosterone treated groups as compared to vehicle control. Cycloheximide 145-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9027330-4 1996 c-fos mRNA levels were decreased 1 h after testosterone treatment in the ventral prostate, whereas they were increased in cycloheximide alone or cycloheximide-testosterone treated groups as compared to vehicle control. Testosterone 159-171 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9027330-5 1996 c-fos protein was also increased in the testosterone-cycloheximide treated group as compared to testosterone alone or cycloheximide alone groups at 1 h. By 3 h, the tissue recovers from the inhibitory effect of cycloheximide as evidenced by restoration of AR and an increase in AR mRNA levels. testosterone-cycloheximide 40-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9027330-5 1996 c-fos protein was also increased in the testosterone-cycloheximide treated group as compared to testosterone alone or cycloheximide alone groups at 1 h. By 3 h, the tissue recovers from the inhibitory effect of cycloheximide as evidenced by restoration of AR and an increase in AR mRNA levels. Testosterone 40-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9027330-5 1996 c-fos protein was also increased in the testosterone-cycloheximide treated group as compared to testosterone alone or cycloheximide alone groups at 1 h. By 3 h, the tissue recovers from the inhibitory effect of cycloheximide as evidenced by restoration of AR and an increase in AR mRNA levels. Cycloheximide 53-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9027330-5 1996 c-fos protein was also increased in the testosterone-cycloheximide treated group as compared to testosterone alone or cycloheximide alone groups at 1 h. By 3 h, the tissue recovers from the inhibitory effect of cycloheximide as evidenced by restoration of AR and an increase in AR mRNA levels. Cycloheximide 118-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9027330-6 1996 At this time c-fos protein levels were reduced after treatment with cycloheximide and testosterone and c-fos mRNA levels were comparable to the controls. Cycloheximide 68-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 8930788-7 1996 (ii) Urethane anaesthesia induced an increase in Fos-like immunoreactivity (FLI) over the basal condition. Urethane 5-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 8982664-0 1996 Lack of cross-tolerance between haloperidol and clozapine towards Fos-protein induction in rat forebrain regions. Haloperidol 32-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 8982664-0 1996 Lack of cross-tolerance between haloperidol and clozapine towards Fos-protein induction in rat forebrain regions. Clozapine 48-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 8982664-1 1996 We investigated whether the acute effects of haloperidol and clozapine on Fos expression in the rat forebrain are mediated by the same receptors through evaluation of cross-tolerance, particularly in the commonly affected areas. Haloperidol 45-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 8982664-1 1996 We investigated whether the acute effects of haloperidol and clozapine on Fos expression in the rat forebrain are mediated by the same receptors through evaluation of cross-tolerance, particularly in the commonly affected areas. Clozapine 61-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 8982664-8 1996 In none of the investigated brain regions was cross-tolerance towards Fos-protein induction found after haloperidol challenge in the clozapine-treated rats. Haloperidol 104-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 8982664-8 1996 In none of the investigated brain regions was cross-tolerance towards Fos-protein induction found after haloperidol challenge in the clozapine-treated rats. Clozapine 133-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 9001738-0 1996 Differential induction of Fos in the female rat brain following different amounts of vaginocervical stimulation: modulation by steroid hormones. Steroids 127-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 9001738-8 1996 These data demonstrate that cells within different estrogen-concentrating regions of the female rat brain are differentially sensitive to VCS, and that steroid hormones can either increase or decrease the amount of Fos induced by different amounts of VCS. Steroids 152-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 215-218 8973831-5 1996 Systemic administration of N-CHA but not of DPMA inhibited light (300 lux, 1 h)-induced Fos expression in the suprachiasmatic nucleus in a dose-dependent manner; 1 mg/kg N-CHA caused 73% inhibition of light-induced Fos expression. N(6)-cyclohexyladenosine 27-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 8973831-5 1996 Systemic administration of N-CHA but not of DPMA inhibited light (300 lux, 1 h)-induced Fos expression in the suprachiasmatic nucleus in a dose-dependent manner; 1 mg/kg N-CHA caused 73% inhibition of light-induced Fos expression. N(6)-cyclohexyladenosine 27-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 215-218 8973831-5 1996 Systemic administration of N-CHA but not of DPMA inhibited light (300 lux, 1 h)-induced Fos expression in the suprachiasmatic nucleus in a dose-dependent manner; 1 mg/kg N-CHA caused 73% inhibition of light-induced Fos expression. N(6)-cyclohexyladenosine 170-175 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 8973831-5 1996 Systemic administration of N-CHA but not of DPMA inhibited light (300 lux, 1 h)-induced Fos expression in the suprachiasmatic nucleus in a dose-dependent manner; 1 mg/kg N-CHA caused 73% inhibition of light-induced Fos expression. N(6)-cyclohexyladenosine 170-175 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 215-218 8945758-2 1996 A single melatonin administration during late day to rats maintained originally under a short photoperiod with 8 h of light per day or under a long photoperiod with 16 h of light per day phase advanced immediately the evening rise in the light-induced SCN c-Fos immunochemistry by about 1.5 h. The data indicate that melatonin administration in vivo may instantaneously reset the intrinsic SCN rhythmicity. Melatonin 9-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 256-261 8955526-0 1996 Fos induction in subtypes of cerebrocortical neurons following single picrotoxin-induced seizures. Picrotoxin 70-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 8981440-0 1996 Ulcerogenic cinchophen induces c-Fos expression in CRH-secreting cells in the PVH of rat brain. cinchophen 12-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 8981440-1 1996 This study aimed at obtaining evidence that cinchophen, an ulcerogenic drug, stimulates the corticotropin-releasing hormone (CRH)-secreting cells in the paraventricular nucleus of the hypothalamus (PVH) to induce c-Fos expression. cinchophen 44-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-218 8981460-1 1996 The distribution of c-fos proto-oncogene expression has been studied in the periaqueductal grey matter (PAG) of non-intentional-stimulated rats by immunohistochemistry. pag 104-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 8957983-4 1996 Lignocaine did not influence either oedema or VTPP, but reduced spinal c-fos expression at 60 min after carrageenan without later effects. Lidocaine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 8957983-5 1996 Bupivacaine induced an increase in VTPP at 30 and 60 min, limitation of oedema at 60 min and a reduction in spinal c-fos expression at 60 and 180 min, but these effects were not present 240 min after carrageenan. Bupivacaine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 8895341-5 1996 These cytokine-dependent inductions of c-fos mRNA levels were not affected by cyclohexamide, but were diminished by actinomycin D pretreatments, consistent with regulation at the transcriptional level. 4-[2-(3,5-dimethyl-2-oxocyclohexyl)-2-hydroxyethyl]piperidine-2,6-dione 78-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 8895341-5 1996 These cytokine-dependent inductions of c-fos mRNA levels were not affected by cyclohexamide, but were diminished by actinomycin D pretreatments, consistent with regulation at the transcriptional level. Dactinomycin 116-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 8903410-8 1996 HGF, phorbol myristate acetate (PMA) and a DG analog, oleylacetylglycerol (OAG), activated the expression of c-jun and c-fos messenger RNAs (mRNAs). Tetradecanoylphorbol Acetate 5-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 8903410-8 1996 HGF, phorbol myristate acetate (PMA) and a DG analog, oleylacetylglycerol (OAG), activated the expression of c-jun and c-fos messenger RNAs (mRNAs). Tetradecanoylphorbol Acetate 32-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 8903410-8 1996 HGF, phorbol myristate acetate (PMA) and a DG analog, oleylacetylglycerol (OAG), activated the expression of c-jun and c-fos messenger RNAs (mRNAs). 1-oleoyl-2-acetylglycerol 54-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 8903410-8 1996 HGF, phorbol myristate acetate (PMA) and a DG analog, oleylacetylglycerol (OAG), activated the expression of c-jun and c-fos messenger RNAs (mRNAs). 1-oleoyl-2-acetylglycerol 75-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 9030697-5 1997 In the LC, PTZ increased mRNA expression of the immediate early gene, c-fos, and mRNA expression of the synthesizing enzyme, tyrosine hydroxylase (TH), and the re-uptake protein, norepinephrine transporter (NET). Pentylenetetrazole 11-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 9038892-4 1997 In in vitro studies, exposure of control tissue to 50 mM K(+)-Krebs-EA (2 x 10(-5) M) solutions significantly increased Fos immunoreactivity in the myenteric plexus, whereas a basal level of Fos was seen in control tissue incubated in Krebs solution, sham-sensitized tissue exposed to bovine serum albumin (BSA, 2 x 10(-5) M), or EA and sensitized tissue exposed to BSA. k(+)-krebs 57-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 191-194 9038892-4 1997 In in vitro studies, exposure of control tissue to 50 mM K(+)-Krebs-EA (2 x 10(-5) M) solutions significantly increased Fos immunoreactivity in the myenteric plexus, whereas a basal level of Fos was seen in control tissue incubated in Krebs solution, sham-sensitized tissue exposed to bovine serum albumin (BSA, 2 x 10(-5) M), or EA and sensitized tissue exposed to BSA. krebs 62-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-123 9038892-4 1997 In in vitro studies, exposure of control tissue to 50 mM K(+)-Krebs-EA (2 x 10(-5) M) solutions significantly increased Fos immunoreactivity in the myenteric plexus, whereas a basal level of Fos was seen in control tissue incubated in Krebs solution, sham-sensitized tissue exposed to bovine serum albumin (BSA, 2 x 10(-5) M), or EA and sensitized tissue exposed to BSA. krebs 62-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 191-194 9038892-5 1997 Pretreatment of sensitized tissue with doxantrazole (10(-4) M) markedly reduced Fos immunoreactivity observed after EA exposure. doxantrazole 39-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 9038892-8 1997 Significantly greater levels of Fos were observed in the myenteric plexus of sensitized animals challenged with EA, even after pretreatment with capsaicin (125 mg/kg). ea 112-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 9038892-8 1997 Significantly greater levels of Fos were observed in the myenteric plexus of sensitized animals challenged with EA, even after pretreatment with capsaicin (125 mg/kg). Capsaicin 145-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 8937722-14 1996 In contrast, haloperidol-induced striatal c-fos mRNA expression was limited to the innervated striatum and was not blocked by SCH-23390. Haloperidol 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 8950089-0 1996 The reinforcing properties of nicotine are associated with a specific patterning of c-fos expression in the rat brain. Nicotine 30-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 8950089-2 1996 The effect of nicotine self-administration on regional brain activity was studied by mapping changes of c-fos expression. Nicotine 14-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 8950089-3 1996 Specific nicotine effects were determine by comparing the patterning of Fos-like immunoreactivity (Fos-Ll) in nicotine self-administering rats with that in three different control groups. Nicotine 9-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 8950089-3 1996 Specific nicotine effects were determine by comparing the patterning of Fos-like immunoreactivity (Fos-Ll) in nicotine self-administering rats with that in three different control groups. Nicotine 9-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 8950089-3 1996 Specific nicotine effects were determine by comparing the patterning of Fos-like immunoreactivity (Fos-Ll) in nicotine self-administering rats with that in three different control groups. Nicotine 110-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 8950089-3 1996 Specific nicotine effects were determine by comparing the patterning of Fos-like immunoreactivity (Fos-Ll) in nicotine self-administering rats with that in three different control groups. Nicotine 110-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 8950089-6 1996 Nicotine self-administration, exposure to saline and food restriction increased Fos-Ll in 43, 33 and three brain regions, respectively, when compared with the control group fed ad libitum. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 8950089-6 1996 Nicotine self-administration, exposure to saline and food restriction increased Fos-Ll in 43, 33 and three brain regions, respectively, when compared with the control group fed ad libitum. Sodium Chloride 42-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 8950089-7 1996 Computer-assisted image analysis of Fos-Ll profiles performed on 16 relevant limbic and sensory structures showed that in saline-treated rats a significant (P < 0.01) increase of Fos-Ll profiles was observed in medial prefrontal cortex, lateral septum, core and ventral shell of nucleus accumbens, claustrum, amygdaloid nuclei, paraventricular thalamic nucleus and lateral geniculate nucleus. Sodium Chloride 122-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-39 8950089-7 1996 Computer-assisted image analysis of Fos-Ll profiles performed on 16 relevant limbic and sensory structures showed that in saline-treated rats a significant (P < 0.01) increase of Fos-Ll profiles was observed in medial prefrontal cortex, lateral septum, core and ventral shell of nucleus accumbens, claustrum, amygdaloid nuclei, paraventricular thalamic nucleus and lateral geniculate nucleus. Sodium Chloride 122-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 182-185 8950098-0 1996 5-HT1A receptor agonist flesinoxan enhances Fos immunoreactivity in rat central amygdala, bed nucleus of the stria terminalis and hypothalamus. flesinoxan 24-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 8950098-5 1996 However, in typically non-5HT1A receptor-containing brain areas Fos-ir is increased due to flesinoxan treatment, as in the paraventricular nucleus of the hypothalamus (PVN), the dorsolateral part of the bed nucleus of the stria terminalis (BNSTdl) and the central amygdala (CeA). flesinoxan 91-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 8950098-8 1996 Moreover, a significant correlation existed between the number of Fos-ir neurons in the PVN and the plasma corticosterone level. Corticosterone 107-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 8933361-9 1996 Our results show that the antagonist NAN-190: (1) completely blocked the photically-induced increase of melatonin receptor density in the SCN, with an IC50 = 0.352 +/- 0.103 mg/kg, and (2) partially blocked (30%) the photic induction of Fos (the protein product of c-fos) in the ventrolateral subdivision of the SCN. 1-(2-methoxyphenyl)-4-(4-(2-phthalimido)butyl)piperazine 37-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 237-240 8933361-9 1996 Our results show that the antagonist NAN-190: (1) completely blocked the photically-induced increase of melatonin receptor density in the SCN, with an IC50 = 0.352 +/- 0.103 mg/kg, and (2) partially blocked (30%) the photic induction of Fos (the protein product of c-fos) in the ventrolateral subdivision of the SCN. 1-(2-methoxyphenyl)-4-(4-(2-phthalimido)butyl)piperazine 37-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 265-270 8933361-10 1996 The agonist 8-OH-DPAT enhanced the photically-induced increase of melatonin receptors by 10% and decreased the photically-induced increase in Fos by 18%. 8-Hydroxy-2-(di-n-propylamino)tetralin 12-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-145 8949925-4 1996 administration of phenylephrine (2.5, 5.0 or 10.0 micrograms/kg) resulted in a significant increase in Fos-like immunoreactivity (Fos-LI), primarily in the caudal part of the NTS. Phenylephrine 18-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-106 8903410-10 1996 Butanol and propranolol at concentrations which effectively inhibited the formation of DG, suppressed HGF-induced expression of c-jun and c-fos mRNAs. Butanols 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 8903410-10 1996 Butanol and propranolol at concentrations which effectively inhibited the formation of DG, suppressed HGF-induced expression of c-jun and c-fos mRNAs. Propranolol 12-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 8916271-3 1996 In other rats that were not allowed to drink after captopril+BK, the induction of Fos-like immunoreactivity (IR) was much lower in the subfornical organ, supraoptic and median preoptic nuclei of old rats compared with their young counterparts. Berkelium 61-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 8898725-0 1996 C-fos mRNA pattern and corticotropin-releasing factor neuronal activity throughout the brain of rats injected centrally with a prostaglandin of E2 type. Prostaglandins 127-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8898725-1 1996 The present study investigated the effect of central administration of the prostaglandin of E2 type (PGE2) on the distribution of the immediate early gene (IEG) c-fos mRNA and the transcriptional activity of corticotropin-releasing factor (CRF) and its type 1 receptor in the brain of conscious rats. Prostaglandins 75-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 8898725-1 1996 The present study investigated the effect of central administration of the prostaglandin of E2 type (PGE2) on the distribution of the immediate early gene (IEG) c-fos mRNA and the transcriptional activity of corticotropin-releasing factor (CRF) and its type 1 receptor in the brain of conscious rats. Dinoprostone 101-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 8898725-11 1996 Taken together, these results provide clear anatomical evidence that central PGE2 injection causes specific and selective expression of c-fos in several brain structures recognized to be activated in the brains of endotoxin-challenged rats. Dinoprostone 77-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 8947933-0 1996 Psychomotor stimulant- and opiate-induced c-fos mRNA expression patterns in the rat forebrain: comparisons between acute drug treatment and a drug challenge in sensitized animals. Opiate Alkaloids 27-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 8947933-1 1996 Amphetamine-, cocaine-, and morphine-induced c-fos expression patterns were examined following an injection protocol that has previously been shown to produce behavioral sensitization and enhanced dopamine release in the striatal complex. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 8947933-1 1996 Amphetamine-, cocaine-, and morphine-induced c-fos expression patterns were examined following an injection protocol that has previously been shown to produce behavioral sensitization and enhanced dopamine release in the striatal complex. Morphine 28-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 8947933-1 1996 Amphetamine-, cocaine-, and morphine-induced c-fos expression patterns were examined following an injection protocol that has previously been shown to produce behavioral sensitization and enhanced dopamine release in the striatal complex. Dopamine 197-205 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 8949925-4 1996 administration of phenylephrine (2.5, 5.0 or 10.0 micrograms/kg) resulted in a significant increase in Fos-like immunoreactivity (Fos-LI), primarily in the caudal part of the NTS. Phenylephrine 18-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-133 8949925-5 1996 This increase in Fos-LI in the barosensitive NTS neurons was appreciably reduced by bilateral microinjection into the caudal NTS of an antisense oligonucleotide (20 pmol, 20 nl) designed to target a region of the c-fos mRNA that flanks the initiation codon (5"-129 to 143-3"). Oligonucleotides 145-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 8949925-5 1996 This increase in Fos-LI in the barosensitive NTS neurons was appreciably reduced by bilateral microinjection into the caudal NTS of an antisense oligonucleotide (20 pmol, 20 nl) designed to target a region of the c-fos mRNA that flanks the initiation codon (5"-129 to 143-3"). Oligonucleotides 145-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-218 8878442-5 1996 Troglitazone (1 microM), a member of the thiazolidinedione class, produced a near complete inhibition of both bFGF-induced DNA synthesis as measured by bromodeoxyuridine incorporation (6.5+/-3.9 vs. 17.6+/-4.3% cells labeled, P < 0.05) and c-fos induction. Troglitazone 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 243-248 8878442-6 1996 This effect was associated with an inhibition (by 73+/-4%, P < 0.01) by troglitazone of the transactivation of the serum response element, which regulates c-fos expression. Troglitazone 75-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 8878442-7 1996 Inhibition of c-fos induction by troglitazone appeared to occur via a blockade of the MAP kinase pathway at a point downstream of MAP kinase activation by MAP kinase kinase. Troglitazone 33-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 8891947-6 1996 Further, c-Fos-like protein in the striatum, considered a marker for the induction of antipsychotic actions by neuroleptic treatments, was downregulated by chronic treatment with the two potent antipsychotic drugs tested, but not by chronic treatment with metoclopramide, which has low antipsychotic efficacy but induces extrapyramidal side effects. Metoclopramide 256-270 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 8922677-2 1996 In an effort to reveal neuronal mechanisms underlying these behavioral difference we determined the effects of the partial D1 agonist SKF 38393, the muscarinic antagonist scopolamine, and the combination of the two drugs on the induction of c-fos in the striatum and its projection sites, the globus pallidus and substantia nigra. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 134-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 241-246 8922677-11 1996 The effects of scopolamine and SKF 38393 on c-fos induction in striatum are qualitatively similar to those reported in rats DA-depleted as adults and suggest that, at this single-label level of analysis, the ability of D1 and muscarinic receptors to influence striatal activity does not contribute to the marked age-related differences in the behavioral effects of DA depletions. Scopolamine 15-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 8922677-11 1996 The effects of scopolamine and SKF 38393 on c-fos induction in striatum are qualitatively similar to those reported in rats DA-depleted as adults and suggest that, at this single-label level of analysis, the ability of D1 and muscarinic receptors to influence striatal activity does not contribute to the marked age-related differences in the behavioral effects of DA depletions. 1,2,3,4-tetrahydroisoquinoline-7-sulfonamide 31-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 8899886-5 1996 Water intake inhibits c-fos in the SON and PVN; whether this is a direct interaction between Ang-II and altered osmolality on magnocellular neurons or a secondary event of an action elsewhere (e.g., in the AV3V) remains uncertain. Water 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 8815937-8 1996 A significant reduction in the number of Fos-like immunoreactive neurons was found ipsilateral to the formalin stimulus in nociresponsive areas of the dorsal horn after on-site injections of morphine into the RAIC. Formaldehyde 102-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 8815937-8 1996 A significant reduction in the number of Fos-like immunoreactive neurons was found ipsilateral to the formalin stimulus in nociresponsive areas of the dorsal horn after on-site injections of morphine into the RAIC. Morphine 191-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 8930328-0 1996 FOS expression induced by interleukin-1 or acute morphine treatment in the rat hypothalamus is attenuated by chronic exposure to morphine. Morphine 49-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 8930328-0 1996 FOS expression induced by interleukin-1 or acute morphine treatment in the rat hypothalamus is attenuated by chronic exposure to morphine. Morphine 129-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 8912249-2 1996 After stimulation of the arterial baroreceptor or the chemoreceptor, we identified c-Fos-labeled neurons with immunoreactions to antisera of glutamate. Glutamic Acid 141-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 8930328-5 1996 We have previously shown that acute treatment with either morphine or IL-1 induces FOS immunoreactivity in the rat brain, including the paraventricular (PVN) and supraoptic (SON) nuclei of the hypothalamus. Morphine 58-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 8930328-6 1996 In this study, using immunocytochemical staining of FOS, we demonstrate that chronic exposure to morphine attenuates the cellular responsiveness to IL-1 and to morphine in the PVN and SON, whereas pretreatment with naloxone, an opiate receptor antagonist, does not reverse the effect of IL-1 on FOS expression. Morphine 97-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 8930328-6 1996 In this study, using immunocytochemical staining of FOS, we demonstrate that chronic exposure to morphine attenuates the cellular responsiveness to IL-1 and to morphine in the PVN and SON, whereas pretreatment with naloxone, an opiate receptor antagonist, does not reverse the effect of IL-1 on FOS expression. Morphine 97-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 295-298 8930328-6 1996 In this study, using immunocytochemical staining of FOS, we demonstrate that chronic exposure to morphine attenuates the cellular responsiveness to IL-1 and to morphine in the PVN and SON, whereas pretreatment with naloxone, an opiate receptor antagonist, does not reverse the effect of IL-1 on FOS expression. Morphine 160-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 8930328-6 1996 In this study, using immunocytochemical staining of FOS, we demonstrate that chronic exposure to morphine attenuates the cellular responsiveness to IL-1 and to morphine in the PVN and SON, whereas pretreatment with naloxone, an opiate receptor antagonist, does not reverse the effect of IL-1 on FOS expression. Morphine 160-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 295-298 8930328-6 1996 In this study, using immunocytochemical staining of FOS, we demonstrate that chronic exposure to morphine attenuates the cellular responsiveness to IL-1 and to morphine in the PVN and SON, whereas pretreatment with naloxone, an opiate receptor antagonist, does not reverse the effect of IL-1 on FOS expression. Naloxone 215-223 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 295-298 8899886-8 1996 dizocilpine (an NMDA open channel antagonist) reduces both drinking and c-fos expression after i.c.v. Dizocilpine Maleate 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 8899888-5 1996 Treatments that induce sodium appetite all induce Fos along the lamina terminalis, but usually not in the SO or PVN. Sodium 23-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 8911658-4 1996 In ovariectomised oil-treated rats, a third ventricular infusion of noradrenaline (45 micrograms) resulted in a significant (P < 0.05) increase in the numbers of Fos-immunoreactive cell nuclei throughout the preoptic area, compared to vehicle controls. Oils 18-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-168 8911658-4 1996 In ovariectomised oil-treated rats, a third ventricular infusion of noradrenaline (45 micrograms) resulted in a significant (P < 0.05) increase in the numbers of Fos-immunoreactive cell nuclei throughout the preoptic area, compared to vehicle controls. Norepinephrine 68-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-168 8911658-9 1996 These results show that noradrenaline-induced Fos expression in the preoptic area is dependent on estrogen status and suggest that the estrogenic regulation of reproductive functions may thus involve altered responses to noradrenaline in sub-populations of preoptic neurones. Norepinephrine 24-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 8873559-0 1996 Halothane and diazepam inhibit ketamine-induced c-fos expression in the rat cingulate cortex. Halothane 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 8873559-0 1996 Halothane and diazepam inhibit ketamine-induced c-fos expression in the rat cingulate cortex. Diazepam 14-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 8873559-0 1996 Halothane and diazepam inhibit ketamine-induced c-fos expression in the rat cingulate cortex. Ketamine 31-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 8873559-2 1996 Recent studies have shown that noncompetitive N-methyl-D-aspartate antagonists cause morphologic damage to the cingulate and retrosplenial cortices and induce c-fos protein (c-Fos) in the same regions. N-Methylaspartate 46-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-164 8873559-2 1996 Recent studies have shown that noncompetitive N-methyl-D-aspartate antagonists cause morphologic damage to the cingulate and retrosplenial cortices and induce c-fos protein (c-Fos) in the same regions. N-Methylaspartate 46-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 174-179 8873559-4 1996 METHODS: The effects of diazepam and halothane on c-Fos expression induced by ketamine were studied. Diazepam 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 8873559-4 1996 METHODS: The effects of diazepam and halothane on c-Fos expression induced by ketamine were studied. Halothane 37-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 8873559-4 1996 METHODS: The effects of diazepam and halothane on c-Fos expression induced by ketamine were studied. Ketamine 78-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 8873559-9 1996 RESULTS: Ketamine induced c-Fos-like immunoreactivity in the cingulate and retrosplenial cortices, thalamus, and neocortex. Ketamine 9-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 8873559-10 1996 Diazepam suppressed the ketamine-induced c-Fos-like immunoreactivity in the cingulate and retrosplenial cortices in a dose-dependent manner, leaving the thalamus and neocortex less affected. Diazepam 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 8873559-10 1996 Diazepam suppressed the ketamine-induced c-Fos-like immunoreactivity in the cingulate and retrosplenial cortices in a dose-dependent manner, leaving the thalamus and neocortex less affected. Ketamine 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 8873559-11 1996 Halothane suppressed the ketamine-induced c-Fos-like immunoreactivity in the cingulate and retrosplenial cortices and the neocortex in a dose-dependent manner, leaving the thalamus relatively unaffected. Halothane 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 8873559-11 1996 Halothane suppressed the ketamine-induced c-Fos-like immunoreactivity in the cingulate and retrosplenial cortices and the neocortex in a dose-dependent manner, leaving the thalamus relatively unaffected. Ketamine 25-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 8873559-12 1996 CONCLUSION: Halothane and diazepam inhibited ketamine-induced c-Fos expression in the cingulate and retrosplenial cortices, leaving the thalamus relatively unaffected. Halothane 12-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 8873559-12 1996 CONCLUSION: Halothane and diazepam inhibited ketamine-induced c-Fos expression in the cingulate and retrosplenial cortices, leaving the thalamus relatively unaffected. Diazepam 26-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 8873559-12 1996 CONCLUSION: Halothane and diazepam inhibited ketamine-induced c-Fos expression in the cingulate and retrosplenial cortices, leaving the thalamus relatively unaffected. Ketamine 45-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 23282563-2 1996 The objectives of this study were to (a) assess the antiinflammatory activity of sulfasalazine (SAZ), a drug known to be effective in the treatment of human UC in a model of chronic granulomatous colitis in rats and (b) determine whether enteral diets supplemented with either FO or two indigestible oligosaccharides (fructooligosaccharide, FOS; xylooligosaccharide, XOS) could attenuate the inflammation observed in a model of chronic granulomatous colitis. Sulfasalazine 81-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 341-344 23282563-2 1996 The objectives of this study were to (a) assess the antiinflammatory activity of sulfasalazine (SAZ), a drug known to be effective in the treatment of human UC in a model of chronic granulomatous colitis in rats and (b) determine whether enteral diets supplemented with either FO or two indigestible oligosaccharides (fructooligosaccharide, FOS; xylooligosaccharide, XOS) could attenuate the inflammation observed in a model of chronic granulomatous colitis. Sulfasalazine 96-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 341-344 23282563-14 1996 The FOS and XOS diets significantly attenuated the PG/PS-induced increases in liver weights when compared with PG/PS + chow-fed animals. pg 51-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-7 23282563-14 1996 The FOS and XOS diets significantly attenuated the PG/PS-induced increases in liver weights when compared with PG/PS + chow-fed animals. Phosphorus 54-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-7 8858922-7 1996 The induction of Fos in granule cells was independent of extracellular calcium ion concentration, but the ryanodine receptor antagonist dantrolene significantly inhibited the response to ETs, suggesting that the mobilisation of calcium ions from intracellular stores is important. Calcium 228-235 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 8884771-6 1996 The removal of circulating androgen by gonadectomy potentiated, whereas dihydrotestosterone treatment of castrates attenuated, the behaviourally induced expression of c-fos messenger RNA in the CA1 region of the hippocampus. Dihydrotestosterone 72-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 167-172 8951924-3 1996 We have examined the time-course of c-fos expression after inflammation produced by either an intra-plantar injection of the irritant turpentine oil or of complete Freund"s adjuvant (CFA). Turpentine 134-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 8951933-0 1996 Ketoprofen produces profound inhibition of spinal c-Fos protein expression resulting from an inflammatory stimulus but not from noxious heat. Ketoprofen 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 8951933-1 1996 This study assesses the anti-inflammatory/analgesic effects of ketoprofen a non-steroidal anti-inflammatory drug, using the method of c-Fos immunoreactivity at the spinal cord level in two models of noxious stimulation: carrageenan-induced inflammatory pain or acute noxious heat. Ketoprofen 63-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-139 8951933-5 1996 dose-dependently blocks the development of the carrageenan-induced spinal c-Fos protein expression and peripheral oedema, with the highest dose influencing in parallel both parameters (75 +/- 2% diminution of total number of c-Fos-LI neurons per L4-L5 section; 64 +/- 4% and 82 +/- 6% diminution of paw and ankle oedema, respectively). Carrageenan 47-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 8951933-5 1996 dose-dependently blocks the development of the carrageenan-induced spinal c-Fos protein expression and peripheral oedema, with the highest dose influencing in parallel both parameters (75 +/- 2% diminution of total number of c-Fos-LI neurons per L4-L5 section; 64 +/- 4% and 82 +/- 6% diminution of paw and ankle oedema, respectively). Carrageenan 47-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 225-230 8951933-6 1996 The effect of ketoprofen was significantly greater on the number of c-Fos-LI neurons in deep, as compared to superficial, laminae. Ketoprofen 14-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 8863847-11 1996 P2Y purinoceptor stimulation with ATP also caused accumulation of c-fos and c-myc mRNAs. Adenosine Triphosphate 34-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 8951933-7 1996 Furthermore, the dose-dependent effects of ketoprofen on the carrageenan-induced spinal c-Fos protein expression and both the paw and ankle oedema were correlated. Ketoprofen 43-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 8951933-7 1996 Furthermore, the dose-dependent effects of ketoprofen on the carrageenan-induced spinal c-Fos protein expression and both the paw and ankle oedema were correlated. Carrageenan 61-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 8951933-8 1996 Oral pre-administration of ketoprofen (20 mg/kg) produced the blockage of development of the carrageenan-induced spinal c-Fos protein expression (65 +/- 3% diminution of total number of c-Fos-LI neurons per L4-L5 section) and peripheral oedema (20 +/- 3% and 59 +/- 10% diminution of paw and ankle oedema, respectively). Ketoprofen 27-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 8951933-8 1996 Oral pre-administration of ketoprofen (20 mg/kg) produced the blockage of development of the carrageenan-induced spinal c-Fos protein expression (65 +/- 3% diminution of total number of c-Fos-LI neurons per L4-L5 section) and peripheral oedema (20 +/- 3% and 59 +/- 10% diminution of paw and ankle oedema, respectively). Ketoprofen 27-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 186-191 8951933-8 1996 Oral pre-administration of ketoprofen (20 mg/kg) produced the blockage of development of the carrageenan-induced spinal c-Fos protein expression (65 +/- 3% diminution of total number of c-Fos-LI neurons per L4-L5 section) and peripheral oedema (20 +/- 3% and 59 +/- 10% diminution of paw and ankle oedema, respectively). Carrageenan 93-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 8951933-8 1996 Oral pre-administration of ketoprofen (20 mg/kg) produced the blockage of development of the carrageenan-induced spinal c-Fos protein expression (65 +/- 3% diminution of total number of c-Fos-LI neurons per L4-L5 section) and peripheral oedema (20 +/- 3% and 59 +/- 10% diminution of paw and ankle oedema, respectively). Carrageenan 93-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 186-191 8951933-11 1996 The method of c-Fos protein-like immunoreactivity revealed ketoprofen to be more potent in comparison to members of other families of non-steroidal anti-inflammatory drugs, previously studied in the same experimental conditions of carrageenan-induced inflammatory pain. Ketoprofen 59-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 8844973-4 1996 In cells loaded with EGTA/AM or treated in low or no Ca2+ HBSS, c-fos induction was reduced similarly in both cell types. Egtazic Acid 21-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 8904794-4 1996 The present work examined the light-induced Fos protein expression on the SCN in the pilocarpine model of chronic epilepsy. Pilocarpine 85-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 8844973-4 1996 In cells loaded with EGTA/AM or treated in low or no Ca2+ HBSS, c-fos induction was reduced similarly in both cell types. ca2+ hbss 53-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 8844973-5 1996 However, inhibition of protein kinases with staurosporin and calphostin C reduced c-fos by 80% in NRK-52E but by only 10-20% in H/1.2NRK.52E. Staurosporine 44-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 8892522-0 1996 c-Fos induction in rat brainstem in response to ethanol- and lithium chloride-induced conditioned taste aversions. Ethanol 48-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8892522-0 1996 c-Fos induction in rat brainstem in response to ethanol- and lithium chloride-induced conditioned taste aversions. Lithium Chloride 61-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8892522-3 1996 c-Fos immunohistochemistry was used to examine neural activation in the rat brainstem associated with drug administration and with a CS taste previously paired with these drugs. Cesium 133-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8892522-4 1996 Relative to saline controls, animals injected with either LiCl (76 mg/kg) or ethanol (3.5 g/kg) displayed greater c-Fos expression in area postrema, nucleus of the solitary tract (NTS), and lateral parabrachial nucleus. Lithium Chloride 58-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 8892522-4 1996 Relative to saline controls, animals injected with either LiCl (76 mg/kg) or ethanol (3.5 g/kg) displayed greater c-Fos expression in area postrema, nucleus of the solitary tract (NTS), and lateral parabrachial nucleus. Ethanol 77-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 8917382-5 1996 The combination of irradiation and beta-adrenergic stimulation by isoproterenol induced earlier expression of jun B and profound expression of the c-fos proto-oncogene in comparison to irradiation alone. Isoproterenol 66-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-152 8917382-8 1996 We observed that the expression of the genes whose regulation is associated with DNA damage (i.e. jun B and c-fos) was enhanced by irradiation alone or in combination with isoproterenol administration. Isoproterenol 172-185 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 8884181-4 1996 GM2 and GM1 inhibit the PDGF-BB-dependent receptor tyrosine autophosphorylation, stimulation of the PLC-gamma 1, increase of inositol-1,4,5-trisphosphate (InsP3), elevation in cytosolic free Ca2+ ([Ca2+]i), expression of the immediate early growth response gene c-fos and cell proliferation with the following rank order of potency GM2 > GM1. gm2 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 262-267 8883820-0 1996 Effect of MK 801 on priming of D1-dependent contralateral turning and its relationship to c-fos expression in the rat caudate-putamen. Dizocilpine Maleate 10-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 8883820-1 1996 In rats with a unilateral 6-hydroxydopamine lesion of the ascending dopamine neurons, we investigated the relationship between the expression of Fos-like immunoreactivity in the caudate-putamen and contralateral turning behavior in response to dopamine agonists during the induction and expression of sensitization (priming) to D1-dependent turning behavior. Oxidopamine 26-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-148 8883820-3 1996 In the induction phase of priming, administration of MK 801 (0.1 mg/kg s.c.) potentiated contralateral turning but differentially influenced stimulation of Fos expression in the caudate-putamen by apomorphine and by SKF 38393. Dizocilpine Maleate 53-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-159 8883820-4 1996 Thus, MK 801 reduced in the expression phase of priming the stimulation of Fos expression by apomorphine in the dorsolateral caudate-putamen, but did not affect that by SKF 38393. Dizocilpine Maleate 6-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 8883820-4 1996 Thus, MK 801 reduced in the expression phase of priming the stimulation of Fos expression by apomorphine in the dorsolateral caudate-putamen, but did not affect that by SKF 38393. Apomorphine 93-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 8883820-6 1996 MK 801, given with apomorphine in the induction phase, reduced the stimulation of Fos expression in the dorsolateral caudate-putamen by SKF 38393. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 8883820-6 1996 MK 801, given with apomorphine in the induction phase, reduced the stimulation of Fos expression in the dorsolateral caudate-putamen by SKF 38393. Apomorphine 19-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 8883820-8 1996 These results are consistent with the possibility that MK 801 disrupts sensitization of D1 transduction by reducing the activation of c-fos by the DA agonist during the induction phase of priming. Dizocilpine Maleate 55-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-139 8756579-2 1996 By Northern blotting of total RNA isolated from PD cells that had been stimulated in the presence of cycloheximide (10 micrograms/ml), PT cell-conditioned medium was shown to induce a significant increase in the expression of the early response gene, c-fos, above both PD cell-conditioned and nonconditioned medium control levels (P < 0.05). Cycloheximide 101-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 251-256 8756579-3 1996 Although forskolin (5 microM) induced a weak increase in c-fos expression in PD cells, the effect of PT medium conditioned in the presence of forskolin enhanced this expression more than additively (P < 0.05); furthermore, this effect was reversed by melatonin. Colforsin 9-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 8884181-4 1996 GM2 and GM1 inhibit the PDGF-BB-dependent receptor tyrosine autophosphorylation, stimulation of the PLC-gamma 1, increase of inositol-1,4,5-trisphosphate (InsP3), elevation in cytosolic free Ca2+ ([Ca2+]i), expression of the immediate early growth response gene c-fos and cell proliferation with the following rank order of potency GM2 > GM1. G(M1) Ganglioside 8-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 262-267 8864900-7 1996 Genistein at 100 micrograms/ml also blocked both bFGF- and PMA-induced increases in c-fos mRNA. Genistein 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 8864900-8 1996 A 24 h pretreatment with PMA at 10(-7) M inhibited the mitogenic response, tyrosine phosphorylation of MAPK, and induction of c-fos mRNA subsequent to the addition of PMA, but not bFGF. Tetradecanoylphorbol Acetate 25-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 8864900-8 1996 A 24 h pretreatment with PMA at 10(-7) M inhibited the mitogenic response, tyrosine phosphorylation of MAPK, and induction of c-fos mRNA subsequent to the addition of PMA, but not bFGF. Tetradecanoylphorbol Acetate 167-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 8877908-1 1996 In the present study we tried to find out whether the competitive NMDA receptor antagonist CGP 40116 was capable of inducing c-Fos expression in the rat cingulate cortex in a manner similar to that described previously for the non-competitive NMDA receptor antagonist MK-801. 2-amino-4-methyl-5-phosphono-3-pentenoic acid 91-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 8877908-5 1996 A qualitatively similar, but quantitatively stronger, effect was observed after administration of MK-801 (0.2 and 0.4 mg/kg), which also caused a dose-dependent increase in the number of c-Fos positive neurones. Dizocilpine Maleate 98-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 187-192 8877908-6 1996 The described dose-dependent effects of CGP 40116 and MK-801 are shown as an increase in the number of c-Fos-positive neurones, but not as an increase in the optical density of c-Fos immunostaining in c-Fos positive neurones. cgp 40-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 8877908-6 1996 The described dose-dependent effects of CGP 40116 and MK-801 are shown as an increase in the number of c-Fos-positive neurones, but not as an increase in the optical density of c-Fos immunostaining in c-Fos positive neurones. Dizocilpine Maleate 54-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 8843090-6 1996 Exposure for 1.5 h to chemosensory cues derived from soiled bedding of estrous females induced Fos immunoreactivity throughout the vomeronasal pathway (i.e. medial amygdala, bed nucleus of the stria terminalis and medial preoptic area) in both ATD and cholesterol males (Experiment 2a). Cholesterol 252-263 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 8890294-2 1996 Intraplantar injection of formalin (5%, 100 microliters in saline) was associated with a high level of spinal c-Fos immunoreactivity and a peripheral paw and ankle edema, as assessed at 3 h after formalin administration. Formaldehyde 26-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 8890294-2 1996 Intraplantar injection of formalin (5%, 100 microliters in saline) was associated with a high level of spinal c-Fos immunoreactivity and a peripheral paw and ankle edema, as assessed at 3 h after formalin administration. Sodium Chloride 59-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 8890294-3 1996 For the two experimental series, the control number of formalin-evoked Fos-like immunoreactive (Fos-LI) neurons were 174 +/- 6 and 193 +/- 18 (means +/- SE) Fos-LI neurons per 40-microns section of the lumbar segment L4-L5 of the rat spinal cord. Formaldehyde 55-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 8890294-3 1996 For the two experimental series, the control number of formalin-evoked Fos-like immunoreactive (Fos-LI) neurons were 174 +/- 6 and 193 +/- 18 (means +/- SE) Fos-LI neurons per 40-microns section of the lumbar segment L4-L5 of the rat spinal cord. Formaldehyde 55-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-99 8890294-3 1996 For the two experimental series, the control number of formalin-evoked Fos-like immunoreactive (Fos-LI) neurons were 174 +/- 6 and 193 +/- 18 (means +/- SE) Fos-LI neurons per 40-microns section of the lumbar segment L4-L5 of the rat spinal cord. Formaldehyde 55-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-99 8890294-8 1996 Laminar analysis of the regional effect of RP67580 on formalin-evoked Fos-LI neurons illustrated that the number of superficial and deep laminae Fos-LI neurons were attenuated to a similar extent by RP67580. Formaldehyde 54-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 8890294-10 1996 Prior intravenous administration of RP68651 (1.5 mg/kg), the inactive isomer of RP67580, produced only a small reduction in the total number of formalin-evoked Fos-LI neurons (84 +/- 5% of the control number of formalin-evoked Fos-LI neurons (P < 0.05). Formaldehyde 144-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-163 8890294-11 1996 The effect of RP68651 on the number of formalin-evoked Fos-LI neurons was significantly smaller (P < 0.01) than the effect of the equivalent concentration of RP67580, the active isomer. Formaldehyde 39-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 8890294-15 1996 Laminar analysis illustrated that coadministered RP67580 and (+)-HA966 reduced the number of formalin-evoked Fos-LI neurons in the superficial and deep laminae to a similar extent. Formaldehyde 93-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-112 8890294-21 1996 The second part of our study suggests that the previously reported NK1/NMDA-receptor interactions contribute to formalin-evoked spinal c-Fos expression and consequently may contribute to the longer term spinal neuroplasticity associated with inflammatory nociceptive processing. Formaldehyde 112-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 9863163-0 1996 Inhibitory effects of dextromethorphan on c-fos protein expression during focal cerebral ischemia in rats. Dextromethorphan 22-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 8883897-0 1996 Nicotine injections into the ventral tegmental area increase locomotion and Fos-like immunoreactivity in the nucleus accumbens of the rat. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 8883897-8 1996 Intra-tegmental injections of nicotine (8.0 micrograms/side) increased Fos-like immunoreactivity in the NAc, but did not affect the number of Fos-positive nuclei in the medial prefrontal cortex or in the dorsolateral striatum. Nicotine 30-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 8883897-9 1996 The increase in accumbal Fos-like immunoreactivity was attenuated by pretreatment with mecamylamine (1.0 mg/kg, s.c.). Mecamylamine 87-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 8939453-4 1996 Brain sections of 30 microns-thickness were made in a cryostat and hybridized with 35S-labelled for c-fos oligonucleotide probe. Sulfur-35 83-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 8939453-7 1996 On the other hand, two typical antipsychotics (haloperidol and fluphenazine) that cause a high incidence of acute motor side effects increased the expression of c-fos mRNA in the dorsolateral striatum, an extrapyramidal region primarily involved in motor control. Haloperidol 47-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 8939453-7 1996 On the other hand, two typical antipsychotics (haloperidol and fluphenazine) that cause a high incidence of acute motor side effects increased the expression of c-fos mRNA in the dorsolateral striatum, an extrapyramidal region primarily involved in motor control. Fluphenazine 63-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 8939453-8 1996 Only clozapine induced c-fos mRNA in the medial prefrontal cortex and lateral septum. Clozapine 5-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 8936476-4 1996 Compared with the single stimulus, repetitive stimulation significantly reduced the number of c-Fos labeled neurons in the VCN by 51%, in the DCN by 75% and in the IC by 48%. polyacrylonitrile 123-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 8840011-8 1996 The study has revealed that, within the time frame associated with the learned aversive response, Fos-immunoreactive (Fos-IR) neurons increased selectively in the central nucleus of the amygdala in animals fed a threonine-imbalanced diet. Threonine 212-221 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 8840011-8 1996 The study has revealed that, within the time frame associated with the learned aversive response, Fos-immunoreactive (Fos-IR) neurons increased selectively in the central nucleus of the amygdala in animals fed a threonine-imbalanced diet. Threonine 212-221 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 8843090-7 1996 By contrast, exposure for 1.5 h to chemosensory cues derived from soiled bedding of sexually active males revealed clear differences between ATD and cholesterol males in neuronal Fos immunoreactive (Experiment 2b). Cholesterol 149-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-182 8897649-0 1996 CO2-induced c-fos expression in the CNS catecholaminergic neurons. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 8758915-7 1996 Induction of MAPK-dependent genes c-fos and MAPK phosphatase-1 by BHTOOH was also blocked by Ras-N-17 expression. 2,6-di-tert-butyl-4-hydroperoxy-4-methyl-2,5-cyclohexadienone 66-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 8809787-3 1996 Electrical intrathecal stimulation (0.5 ms pulses, 15 V, 3 Hz for 15 min) or intrathecal injection of N-methyl-D-aspartate (25 nmol) produced massive c-Fos immunoreactivity in neurons throughout the sacral spinal cord and the dorsal horn of the lumber spinal cord. N-Methylaspartate 102-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-155 8880857-0 1996 A nitric oxide synthesis inhibitor (L-NAME) reduces licking behavior and Fos-labeling in the spinal cord of rats during formalin-induced inflammation. Nitric Oxide 2-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-76 8880857-0 1996 A nitric oxide synthesis inhibitor (L-NAME) reduces licking behavior and Fos-labeling in the spinal cord of rats during formalin-induced inflammation. NG-Nitroarginine Methyl Ester 36-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-76 8880857-4 1996 In addition, these same doses of L-NAME reduced formalin-induced Fos-labeling in the ipsilateral dorsal gray matter (as compared to the contralateral gray matter). NG-Nitroarginine Methyl Ester 33-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 8880857-4 1996 In addition, these same doses of L-NAME reduced formalin-induced Fos-labeling in the ipsilateral dorsal gray matter (as compared to the contralateral gray matter). Formaldehyde 48-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 8855514-0 1996 Amphetamine-induced c-fos mRNA expression is altered in rats with neonatal ventral hippocampal damage. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 8855514-8 1996 These results indicate that the neonatal VH lesion alters time-dependent intracellular signal transduction mechanisms measured by AMPH-induced c-fos mRNA expression in cortical and subcortical brain regions. Amphetamine 130-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-148 8864481-0 1996 Effect of chronic dexfenfluramine on Fos in rat brain. Dexfenfluramine 18-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 8864301-0 1996 Expression of Fos in rat brain in relation to sodium appetite: furosemide and cerebroventricular renin. Sodium 46-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 8864301-0 1996 Expression of Fos in rat brain in relation to sodium appetite: furosemide and cerebroventricular renin. Furosemide 63-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 8864301-1 1996 Two experiments were performed to investigate the relationship between the expression of sodium appetite and the appearance of Fos-like immunoreactivity (Fos-IR) in the brain of rats. Sodium 89-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-130 8864301-1 1996 Two experiments were performed to investigate the relationship between the expression of sodium appetite and the appearance of Fos-like immunoreactivity (Fos-IR) in the brain of rats. Sodium 89-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-157 8864045-5 1996 Patterns of fos-like immunoreactivity (FLI) were observed throughout the brainstem following electrical stimulation of the SC in Urethane-anaesthetized rats. Urethane 129-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-15 8754788-7 1996 The stimulation of TH mRNA was mediated by the AT1 receptor subtype, resulted from an increase in its transcription, and involved activation of phospholipase C and protein kinase C. Antisense oligonucleotide for c-fos attenuated Ang II stimulation of TH mRNA in a time- and dose-dependent fashion, indicating an involvement of c-fos as a putative third messenger in Ang II stimulation of TH. Oligonucleotides 192-207 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 212-217 8754788-7 1996 The stimulation of TH mRNA was mediated by the AT1 receptor subtype, resulted from an increase in its transcription, and involved activation of phospholipase C and protein kinase C. Antisense oligonucleotide for c-fos attenuated Ang II stimulation of TH mRNA in a time- and dose-dependent fashion, indicating an involvement of c-fos as a putative third messenger in Ang II stimulation of TH. Oligonucleotides 192-207 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 327-332 8897649-2 1996 Breathing a gas mixture with elevated CO2 (15% CO2, 21% O2 and 64% N2, or 15% CO2 balance O2) for 60 min, induced activation of the c-fos gene in widespread regions of the CNS, as indicated by the expression of Fos-like immunoreactive protein (Fos). N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 8897649-2 1996 Breathing a gas mixture with elevated CO2 (15% CO2, 21% O2 and 64% N2, or 15% CO2 balance O2) for 60 min, induced activation of the c-fos gene in widespread regions of the CNS, as indicated by the expression of Fos-like immunoreactive protein (Fos). N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 211-214 8897649-2 1996 Breathing a gas mixture with elevated CO2 (15% CO2, 21% O2 and 64% N2, or 15% CO2 balance O2) for 60 min, induced activation of the c-fos gene in widespread regions of the CNS, as indicated by the expression of Fos-like immunoreactive protein (Fos). N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 244-247 8897649-2 1996 Breathing a gas mixture with elevated CO2 (15% CO2, 21% O2 and 64% N2, or 15% CO2 balance O2) for 60 min, induced activation of the c-fos gene in widespread regions of the CNS, as indicated by the expression of Fos-like immunoreactive protein (Fos). N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 47-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 8897649-2 1996 Breathing a gas mixture with elevated CO2 (15% CO2, 21% O2 and 64% N2, or 15% CO2 balance O2) for 60 min, induced activation of the c-fos gene in widespread regions of the CNS, as indicated by the expression of Fos-like immunoreactive protein (Fos). Oxygen 39-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 8897649-2 1996 Breathing a gas mixture with elevated CO2 (15% CO2, 21% O2 and 64% N2, or 15% CO2 balance O2) for 60 min, induced activation of the c-fos gene in widespread regions of the CNS, as indicated by the expression of Fos-like immunoreactive protein (Fos). Nitrogen 67-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 8897649-2 1996 Breathing a gas mixture with elevated CO2 (15% CO2, 21% O2 and 64% N2, or 15% CO2 balance O2) for 60 min, induced activation of the c-fos gene in widespread regions of the CNS, as indicated by the expression of Fos-like immunoreactive protein (Fos). N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 47-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 8897649-2 1996 Breathing a gas mixture with elevated CO2 (15% CO2, 21% O2 and 64% N2, or 15% CO2 balance O2) for 60 min, induced activation of the c-fos gene in widespread regions of the CNS, as indicated by the expression of Fos-like immunoreactive protein (Fos). Oxygen 48-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 8897649-4 1996 Colocalization studies of tyrosine hydroxylase (TH) and Fos protein revealed that in the brainstem, 73 to 85% of noradrenaline-containing cells expressed Fos immunoreactivity. Norepinephrine 113-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 8897649-4 1996 Colocalization studies of tyrosine hydroxylase (TH) and Fos protein revealed that in the brainstem, 73 to 85% of noradrenaline-containing cells expressed Fos immunoreactivity. Norepinephrine 113-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-157 8706827-6 1996 Moreover, we show that c-Fos is immunologically detected in lysosomes isolated from the liver of rats treated with the protease inhibitor leupeptin. leupeptin 138-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 8809832-5 1996 The present study was designed to document the long-term expression (up to seven months) of the fos-related antigens and to map their distributions in the rat brain after systemic treatment with kainic acid. Kainic Acid 195-206 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-99 8809832-12 1996 The fos-related antigens and activator protein-1 binding activity were continuously expressed at high levels throughout the experimental period in the dentate granule cells where mossy fibre collateral sprouting occurred after kainic acid treatment. Kainic Acid 227-238 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-7 8864301-3 1996 Some rats had access to distilled water, and others had no fluids available during the 24 h. All of the furosemide-treated rats showed Fos-IR in both the subfornical organ (SFO) and around the organum vasculosum laminae terminalis (OVLT). Furosemide 104-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-138 8864301-7 1996 Both Fos-IR and fluid intake were antagonized by administration of losartan, an angiotensin II type 1 receptor antagonist. Losartan 67-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 5-8 8864302-4 1996 Cortical Fos expression following amphetamine showed the same general pattern, and was blocked by either a selective D1 or D2 antagonist. Amphetamine 34-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 8842399-0 1996 Dopaminergic transplants suppress L-DOPA-induced Fos expression in the dopamine-depleted striatum in a rat model of Parkinson"s disease. Levodopa 34-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 8842399-0 1996 Dopaminergic transplants suppress L-DOPA-induced Fos expression in the dopamine-depleted striatum in a rat model of Parkinson"s disease. Dopamine 71-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 8842399-9 1996 The stimulatory effect of L-DOPA on c-Fos expression observed within the lesioned striatum was suppressed by fetal VM transplants. Levodopa 26-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 8842399-11 1996 Taken together, these findings suggest that glutamatergic modulation is involved in the L-DOPA-induced c-Fos expression in the denervated striatum which is normalized by fetal VM transplants. Levodopa 88-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 8679218-5 1996 Our results are unique in that they demonstrate that asbestos induces apoptosis in mesothelial cells at concentrations eliciting increased expression of the proto-oncogenes c-fos and c-jun. Asbestos 53-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 173-178 8804719-3 1996 In this study, AP-1 DNA binding activity, an indicator of the functional form of the c-fos transcription factor, was examined in nuclear extracts prepared from these brain regions using an electrophoretic mobility shift assay and a labeled oligonucleotide containing the AP-1 consensus sequence. Oligonucleotides 240-255 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 8670682-1 1996 The present studies compared the effects of acute and chronic administration of haloperidol or clozapine on c-fos mRNA expression in the rat medial prefrontal cortex. Haloperidol 80-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 8670682-1 1996 The present studies compared the effects of acute and chronic administration of haloperidol or clozapine on c-fos mRNA expression in the rat medial prefrontal cortex. Clozapine 95-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 8670682-2 1996 Acute administration of clozapine, but not haloperidol robustly increased c-fos mRNA expression in the infralimbic and prelimbic cortex of the rat. Clozapine 24-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 8670682-3 1996 Even though most c-fos mRNA-expressing neurons in the clozapine- treated animals were localized in deep cortical layers, labeled neurons were found organized into several cell bridges connecting the superficial and deep layers of the cortex. Clozapine 54-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 8670682-4 1996 After chronic treatment with clozapine, c-fos mRNA was reduced by approximately 60% of that seen acutely; however, the columns of c-fos mRNA expressing neurons did not show the same magnitude of tolerance. Clozapine 29-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 8670682-7 1996 Remoxipride, a selective D2 antagonist in vitro, induced c-fos mRNA at very low doses and lost its ability to alter c-fos mRNA at higher doses. Remoxipride 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 8670682-7 1996 Remoxipride, a selective D2 antagonist in vitro, induced c-fos mRNA at very low doses and lost its ability to alter c-fos mRNA at higher doses. Remoxipride 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 8670682-8 1996 Interestingly, U-99194A, an antagonist with 20-fold selectivity for D3 over D2 receptors, also produced greater induction of c-fos mRNA at lower doses. (5,6-dimethoxyindan-2-yl)dipropylamine 15-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 8753884-3 1996 We find that in vivo, the NMDA receptor antagonist MK-801 inhibits amphetamine induction of c-fos acutely and also prevents downregulation of IEG expression with chronic amphetamine administration. Dizocilpine Maleate 51-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 8753884-3 1996 We find that in vivo, the NMDA receptor antagonist MK-801 inhibits amphetamine induction of c-fos acutely and also prevents downregulation of IEG expression with chronic amphetamine administration. Amphetamine 67-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 8965094-4 1996 In the present study, we investigated the histopathological changes in rat brains induced by an intracerebral microinjection of kainic acid, a potent analogue of glutamate using two newly available markers for ischemic neurons: Fos immunohistochemistry and EA 50 stain. Kainic Acid 128-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 228-231 8764375-2 1996 Three hours after the intraplantar injection of carrageenin (6 mg/150 microliters of saline) Fos-like immunoreactivity (Fos-LI) was observed in both superficial and deep laminae of the dorsal horn segments L4 and L5 of the spinal cord. Carrageenan 48-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 8764375-2 1996 Three hours after the intraplantar injection of carrageenin (6 mg/150 microliters of saline) Fos-like immunoreactivity (Fos-LI) was observed in both superficial and deep laminae of the dorsal horn segments L4 and L5 of the spinal cord. Carrageenan 48-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-123 8764375-2 1996 Three hours after the intraplantar injection of carrageenin (6 mg/150 microliters of saline) Fos-like immunoreactivity (Fos-LI) was observed in both superficial and deep laminae of the dorsal horn segments L4 and L5 of the spinal cord. Sodium Chloride 85-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 8764375-2 1996 Three hours after the intraplantar injection of carrageenin (6 mg/150 microliters of saline) Fos-like immunoreactivity (Fos-LI) was observed in both superficial and deep laminae of the dorsal horn segments L4 and L5 of the spinal cord. Sodium Chloride 85-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-123 8764375-11 1996 Both atipamezole and a combined injection of atipamezole and naloxone blocked the effects of medetomidine plus morphine on both the total number of Fos-LI neurons (86 +/- 11% and 86 +/- 6% of control, respectively) and carrageenin inflammation (87 +/- 6%, P < .05 and 84 +/- 3%, P < .05 of control for the paw edema; 75 +/- 8%, P < .01 and 81 +/- 7%, P < .05 of control for the ankle edema, respectively). atipamezole 5-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-151 8764375-11 1996 Both atipamezole and a combined injection of atipamezole and naloxone blocked the effects of medetomidine plus morphine on both the total number of Fos-LI neurons (86 +/- 11% and 86 +/- 6% of control, respectively) and carrageenin inflammation (87 +/- 6%, P < .05 and 84 +/- 3%, P < .05 of control for the paw edema; 75 +/- 8%, P < .01 and 81 +/- 7%, P < .05 of control for the ankle edema, respectively). atipamezole 45-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-151 8764375-11 1996 Both atipamezole and a combined injection of atipamezole and naloxone blocked the effects of medetomidine plus morphine on both the total number of Fos-LI neurons (86 +/- 11% and 86 +/- 6% of control, respectively) and carrageenin inflammation (87 +/- 6%, P < .05 and 84 +/- 3%, P < .05 of control for the paw edema; 75 +/- 8%, P < .01 and 81 +/- 7%, P < .05 of control for the ankle edema, respectively). Naloxone 61-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-151 8764375-11 1996 Both atipamezole and a combined injection of atipamezole and naloxone blocked the effects of medetomidine plus morphine on both the total number of Fos-LI neurons (86 +/- 11% and 86 +/- 6% of control, respectively) and carrageenin inflammation (87 +/- 6%, P < .05 and 84 +/- 3%, P < .05 of control for the paw edema; 75 +/- 8%, P < .01 and 81 +/- 7%, P < .05 of control for the ankle edema, respectively). Medetomidine 93-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-151 8764375-11 1996 Both atipamezole and a combined injection of atipamezole and naloxone blocked the effects of medetomidine plus morphine on both the total number of Fos-LI neurons (86 +/- 11% and 86 +/- 6% of control, respectively) and carrageenin inflammation (87 +/- 6%, P < .05 and 84 +/- 3%, P < .05 of control for the paw edema; 75 +/- 8%, P < .01 and 81 +/- 7%, P < .05 of control for the ankle edema, respectively). Morphine 111-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-151 8764375-12 1996 Naloxone alone blocked the effects of the co-administered agonists on the total number of Fos-Li neurons (91 +/- 6% of the control carrageenin group) without influencing the effect on the peripheral edema. Naloxone 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 8837013-4 1996 A transient induction of both c-fos and c-jun mRNAs by TPA was observed in both cell populations, together with an associated suppression of BSP mRNA in the fetal rat calvarial cells. Tetradecanoylphorbol Acetate 55-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 8865104-3 1996 We hypothesized that butyrate decreases and deoxycholate increases crypt surface proliferation in vivo and that these effects are mediated by changes in the expression of the protooncogenes c-Fos and c-Jun, which are known to regulate proliferation and differentiation. Butyrates 21-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 190-195 8865104-9 1996 Butyrate increased colonic expression of c-Jun, whereas deoxycholate significantly induced c-Fos. Deoxycholic Acid 56-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 8783264-1 1996 In this pharmacological study we have assessed the effect of baclofen, a selective GABAB receptor agonist, on spinal expression of the immediate early gene c-Fos and the peripheral oedema evoked by a prolonged peripheral inflammation due to intraplantar carrageenan. Baclofen 61-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 10456073-1 1996 Recently, we have described a potential neuronal correlate of the behavioral expression of a conditioned taste aversion (CTA) against sucrose at the level of c-Fos expression. Sucrose 134-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 10456073-2 1996 Intraoral infusions of sucrose induce c-Fos-like immunoreactivity (c-FLI) in the intermediate nucleus of the solitary tract (iNTS) after a CTA has been acquired for sucrose. Sucrose 23-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 10456074-1 1996 Local microinjection into rat amygdala of phosphorothioate modified oligodeoxynucleotides (ODNs) antisense to c-fos several hours before conditioned taste aversion (CTA) training impaired taste aversion memory tested 3-5 days after conditioning. Parathion 42-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 10456074-1 1996 Local microinjection into rat amygdala of phosphorothioate modified oligodeoxynucleotides (ODNs) antisense to c-fos several hours before conditioned taste aversion (CTA) training impaired taste aversion memory tested 3-5 days after conditioning. Oligodeoxyribonucleotides 68-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 10456074-1 1996 Local microinjection into rat amygdala of phosphorothioate modified oligodeoxynucleotides (ODNs) antisense to c-fos several hours before conditioned taste aversion (CTA) training impaired taste aversion memory tested 3-5 days after conditioning. Oligodeoxyribonucleotides 91-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 8710367-3 1996 Fos binding sites were immunoselected from random sequence oligonucleotides using a pan Fos anti-serum with nuclear protein from quiescent FBRp75v-fos-transformed (FBR) and normal (208F) rat fibroblasts. Oligonucleotides 59-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 8783238-6 1996 Administration of d-amphetamine increased c-fos expression in the neostriatum and the globus pallidus of the control group. Dextroamphetamine 18-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 8783238-10 1996 Pre-treatment with SCH 23390 blocked the effect of d-amphetamine on c-fos expression in control and grafted animals. SCH 23390 19-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 8783238-10 1996 Pre-treatment with SCH 23390 blocked the effect of d-amphetamine on c-fos expression in control and grafted animals. Dextroamphetamine 51-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 8783238-13 1996 Grafts made into neonates, when challenged with amphetamine, induce an abnormal c-fos expression which can predict the degree of overshoot observed for rotation activity. Amphetamine 48-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 8783246-0 1996 N-methyl-D-aspartate and non-N-methyl-D-aspartate receptor antagonism reduces Fos-like immunoreactivity in central trigeminal neurons after corneal stimulation in the rat. N-Methylaspartate 0-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 8783264-4 1996 For the two series of experiments the total number of control carrageenan-evoked c-Fos protein-like immunoreactive neurons in segments L4-L5 of the spinal cord was 176 +/- 6 and 177 +/- 9 c-Fos protein-like immunoreactive neurons per section, for carrageenan control with intravenous and intraplantar saline, respectively. Carrageenan 62-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 8783264-4 1996 For the two series of experiments the total number of control carrageenan-evoked c-Fos protein-like immunoreactive neurons in segments L4-L5 of the spinal cord was 176 +/- 6 and 177 +/- 9 c-Fos protein-like immunoreactive neurons per section, for carrageenan control with intravenous and intraplantar saline, respectively. Carrageenan 62-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 188-193 8783264-5 1996 c-Fos protein-like immunoreactive neurons were predominantly located in laminae I-II and V-VI of the dorsal horn of the spinal cord in carrageenan controls receiving intravenous (68 +/- 3 and 69 +/- 2 c-Fos protein-like immunoreactive neurons, respectively) and intraplantar (62 +/- 4 and 71 +/- 5 c-Fos protein-like immunoreactive neurons, respectively) saline. Sodium Chloride 355-361 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8783264-8 1996 The effects of systemic baclofen on the carrageenan-induced spinal c-Fos expression and both the paw and ankle oedema were positively correlated (r = 0.479, P < 0.05 and r = 0.733, P < 0.001, respectively). Baclofen 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 8783264-8 1996 The effects of systemic baclofen on the carrageenan-induced spinal c-Fos expression and both the paw and ankle oedema were positively correlated (r = 0.479, P < 0.05 and r = 0.733, P < 0.001, respectively). Carrageenan 40-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 8670150-6 1996 Conjugated and unconjugated bile acids rapidly induced egr and fos gene expression as well as cytoplasmic mitogen-activated protein kinase (MAPK) activation. Bile Acids and Salts 28-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 8807746-4 1996 We therefore examined the effects of administration of two D2 receptor antagonists, haloperidol and metoclopramide, on Fos expression in the striatum and temporal cortices of the vervet monkey. Haloperidol 84-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-122 8807746-4 1996 We therefore examined the effects of administration of two D2 receptor antagonists, haloperidol and metoclopramide, on Fos expression in the striatum and temporal cortices of the vervet monkey. Metoclopramide 100-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-122 8807746-7 1996 Haloperidol but not metoclopramide increased the number of Fos-li neurons in the nucleus accumbens shell. Haloperidol 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 8807746-8 1996 Similarly, haloperidol but not metoclopramide increased the number of Fos-li neurons in the entorhinal cortex. Haloperidol 11-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 8654580-3 1996 The Fos and Jun components of AP1 were induced rapidly and transiently in PC12 cells following the addition of phorbol ester (phorbol 12-myristate 13-acetate, PMA). Phorbol Esters 111-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-7 8828587-6 1996 MK-801 treatment also enhanced striatal c-fos expression produced by A-85653 but only if MK-801 were given in combination with A-85653 2 h prior to sacrifice; prior daily treatment with MK-801 had no carry-over effect. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 8805122-0 1996 Fos oncoprotein expression in the rat forebrain following muscimol-induced absence seizures. Muscimol 58-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 8805122-3 1996 Six hours after a systemic injection of muscimol a massive Fos immunoreactivity appeared in the olfactory system, retrosplenial cortex and paraventricular thalamic nucleus, whereas other cortical areas contained low level of Fos expression. Muscimol 40-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 8805122-3 1996 Six hours after a systemic injection of muscimol a massive Fos immunoreactivity appeared in the olfactory system, retrosplenial cortex and paraventricular thalamic nucleus, whereas other cortical areas contained low level of Fos expression. Muscimol 40-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 225-228 8805122-4 1996 These results provide the first functional morphological evidence suggesting that these forebrain structures with Fos expression may play an important role in the pathophysiology of muscimol-induced absence seizures. Muscimol 182-190 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 8764271-4 1996 High CPP (140 cmH2O), which increased coronary flow (8.99 vs. 17.6 ml/min) and left ventricular systolic pressure (50 vs. 91 mmHg), increased steady state c-fos mRNA expression 2.3-fold (all P < 0.01 vs. low CPP). cmh2o 14-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-160 8856738-0 1996 Systemic application of pyrethroid insecticides evokes differential expression of c-Fos and c-Jun proteins in rat brain. Pyrethrins 24-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 8856738-1 1996 Expression of the c-Fos and c-Jun transcription factor was investigated by immunocytochemistry in the thalamus, hypothalamus, hippocampus and cortex of adult rats following intraperitoneal application of proconvulsant doses of the pyrethroid insecticides, cypermethrin and permethrin. Pyrethrins 231-241 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 8856738-1 1996 Expression of the c-Fos and c-Jun transcription factor was investigated by immunocytochemistry in the thalamus, hypothalamus, hippocampus and cortex of adult rats following intraperitoneal application of proconvulsant doses of the pyrethroid insecticides, cypermethrin and permethrin. cypermethrin 256-268 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 8856738-1 1996 Expression of the c-Fos and c-Jun transcription factor was investigated by immunocytochemistry in the thalamus, hypothalamus, hippocampus and cortex of adult rats following intraperitoneal application of proconvulsant doses of the pyrethroid insecticides, cypermethrin and permethrin. Permethrin 258-268 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 8856738-3 1996 Cypermethrin induced a strong expression of c-Fos and c-Jun in all the thalamic nuclei, except the ventro-posterior complex and substantia nigra, and in all the hypothalamic nuclei. cypermethrin 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 8856738-6 1996 In the hippocampus, cypermethrin induced a weak expression of c-Fos, but not of c-Jun, in the dentate gyrus and CA-3 area. cypermethrin 20-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 8856738-7 1996 Permethrin that has a lower pharmacological potency, evoked a similar pattern of c-Fos and c-Jun expression, however, intensity and persistence of the neuronal labeling were less pronounced. Permethrin 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 8856738-8 1996 Our results demonstrate that the neurotoxic effects of pyrethroid insecticides comprise molecular genetic alterations in the brain such as early and lasting induction of Fos and Jun transcription factor proteins. Pyrethrins 55-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-173 8654580-3 1996 The Fos and Jun components of AP1 were induced rapidly and transiently in PC12 cells following the addition of phorbol ester (phorbol 12-myristate 13-acetate, PMA). Tetradecanoylphorbol Acetate 126-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-7 8654580-3 1996 The Fos and Jun components of AP1 were induced rapidly and transiently in PC12 cells following the addition of phorbol ester (phorbol 12-myristate 13-acetate, PMA). Tetradecanoylphorbol Acetate 159-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-7 8762093-0 1996 Intraplantar morphine depresses spinal c-Fos expression induced by carrageenin inflammation but not by noxious heat. Morphine 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 8762093-0 1996 Intraplantar morphine depresses spinal c-Fos expression induced by carrageenin inflammation but not by noxious heat. Carrageenan 67-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 8762093-4 1996 Intraplantar carrageenin, in awake rats, induced numerous Fos-like immunoreactive (Fos-LI) neurones in the dorsal horn of L4-L5 lumbar segments of the spinal cord and extensive peripheral oedema. Carrageenan 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 8762093-4 1996 Intraplantar carrageenin, in awake rats, induced numerous Fos-like immunoreactive (Fos-LI) neurones in the dorsal horn of L4-L5 lumbar segments of the spinal cord and extensive peripheral oedema. Carrageenan 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 8762093-7 1996 Intraplantar morphine dose-dependently reduced c-Fos expression induced 1 h 30 min after carrageenin (r = 0.605, P < 0.02), these effects were completely blocked by intraplantar methiodide naloxone (20 micrograms) (121 +/- 22% of control carrageenin expression). Morphine 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 8762093-7 1996 Intraplantar morphine dose-dependently reduced c-Fos expression induced 1 h 30 min after carrageenin (r = 0.605, P < 0.02), these effects were completely blocked by intraplantar methiodide naloxone (20 micrograms) (121 +/- 22% of control carrageenin expression). Carrageenan 89-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 8762093-7 1996 Intraplantar morphine dose-dependently reduced c-Fos expression induced 1 h 30 min after carrageenin (r = 0.605, P < 0.02), these effects were completely blocked by intraplantar methiodide naloxone (20 micrograms) (121 +/- 22% of control carrageenin expression). methyl iodide 181-191 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 8641204-6 1996 Ang II stimulation of PAI-1 mRNA succeeded its action on c-fos mRNA and was attenuated by c-fos antisense oligonucleotide. Oligonucleotides 106-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 8799578-0 1996 Tyrphostin inhibition of ATP-stimulated DNA synthesis, cell proliferation and fos-protein expression in vascular smooth muscle cells. Tyrphostins 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 8799578-0 1996 Tyrphostin inhibition of ATP-stimulated DNA synthesis, cell proliferation and fos-protein expression in vascular smooth muscle cells. Adenosine Triphosphate 25-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 8799578-10 1996 When RASMC were incubated with 10(-5) M ATP for 2 h, nearly all of the cells (87 +/- 5%) were intensely stained with an antibody to the Fos protein while in the controls only 1 +/- 2% of the cells were weakly stained. rasmc 5-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-139 8799578-10 1996 When RASMC were incubated with 10(-5) M ATP for 2 h, nearly all of the cells (87 +/- 5%) were intensely stained with an antibody to the Fos protein while in the controls only 1 +/- 2% of the cells were weakly stained. Adenosine Triphosphate 40-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-139 8799578-11 1996 Tyrphostin 25 greatly reduced the Fos-protein staining (14 +/- 2%). Tyrphostins 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 8799578-18 1996 In conclusion, tyrphostin 25 inhibited ATP-induced DNA synthesis, cell proliferation and Fos-protein expression, but not ATP-induced 45Ca(2+)-influx, inositolphosphate-production or vasoconstriction. Tyrphostins 15-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 8762093-7 1996 Intraplantar morphine dose-dependently reduced c-Fos expression induced 1 h 30 min after carrageenin (r = 0.605, P < 0.02), these effects were completely blocked by intraplantar methiodide naloxone (20 micrograms) (121 +/- 22% of control carrageenin expression). Naloxone 192-200 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 8762093-11 1996 In the second series of experiments, intraplantar morphine dose-dependently reduced the number of superficial and deep Fos-LI neurones induced 3 h after carrageenin (r = 0.794, P < 0.0004 and r = 0.698, P < 0.004, respectively). Morphine 50-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-122 8762093-16 1996 Brief noxious heat stimulation, in urethane anaesthetized rats, induced, 2 h after the stimulation, numerous Fos-LI neurones in the dorsal horn of L3-L4 lumbar segments of the spinal cord but no detectable peripheral oedema. Urethane 35-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-112 8752587-3 1996 Intense potentiation of D1-dependent turning behaviour and c-Fos expression was also observed after administration of the A2a/A1 antagonist CGS 15943. cysteinylglycine 140-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 8782081-8 1996 In GH3B6 cells stimulated with TRH or KCl, enhanced FOS protein levels were detected by immunofluorescence and localized in the nucleus with confocal microscopy. Potassium Chloride 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 8752587-4 1996 Administration of the A1 adenosine receptor antagonist DPCPX induced a small potentiation of D1-mediated contralateral turning while c-Fos expression induced by SKF 38393 was not modified. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 161-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 8782081-2 1996 The induction of c-fos mRNA as detected by Northern blot analysis was stimulated by TRH and by depolarization with KCl, both leading to a rise in cytosolic free [Ca2+] ([Ca2+]i), and also by epidermal growth factor (EGF). Potassium Chloride 115-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 8782081-6 1996 c-fos mRNA induction by TRH and KCl was markedly inhibited by two blockers of Ca2+/calmodulin-dependent protein kinase (CaM kinase), KN-62 and calmidazolium. Potassium Chloride 32-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8782081-6 1996 c-fos mRNA induction by TRH and KCl was markedly inhibited by two blockers of Ca2+/calmodulin-dependent protein kinase (CaM kinase), KN-62 and calmidazolium. KN 62 133-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8782081-6 1996 c-fos mRNA induction by TRH and KCl was markedly inhibited by two blockers of Ca2+/calmodulin-dependent protein kinase (CaM kinase), KN-62 and calmidazolium. calmidazolium 143-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8782081-7 1996 In contrast, KCl induction of c-fos and the effects of KN-62 on TRH induction of c-fos were not observed in a closely related pituitary line GH4C1 in which TRH exerts its effects on immediate early genes predominantly via the protein kinase C pathway. Potassium Chloride 13-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 8752587-0 1996 Blockade of A2a adenosine receptors positively modulates turning behaviour and c-Fos expression induced by D1 agonists in dopamine-denervated rats. Dopamine 122-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 8752587-2 1996 SCH 58261 also increased the number of Fos-like positive nuclei induced by SKF 38393 in the 6-hydroxydopamine-lesioned striatum. Oxidopamine 92-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 8861055-0 1996 Scopolamine augments c-fos and zip/268 messenger RNA expression induced by the full D(1) dopamine receptor agonist SKF-82958 in the intact rat striatum. Scopolamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 8861055-5 1996 However, scopolamine attenuated basal, and SKF-82958-stimulated, expression of c-fos and zif/268 messenger RNAs in the cortex. Scopolamine 9-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 8861055-6 1996 Scopolamine also enabled SKF-38393 to induce locomotor stimulation and c-fos and zif/268 messenger RNA expression in the normosensitive striatum of the rat when SKF-38393 alone caused no such changes. Scopolamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 8861055-6 1996 Scopolamine also enabled SKF-38393 to induce locomotor stimulation and c-fos and zif/268 messenger RNA expression in the normosensitive striatum of the rat when SKF-38393 alone caused no such changes. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 25-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 8793085-2 1996 Most granule cells displaying high levels of Fos accumulation are located in the bulbar columns defined by the odour-induced foci of high 2-deoxyglucose glomerular uptake. Deoxyglucose 138-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 8793085-6 1996 It is concluded that c-fos expression induced by a sustained stimulation with propionic acid vapours is not only determined by the olfactory peripheral input but also by afferents of central origin. propionic acid 78-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 8793085-8 1996 The beta-adrenergic antagonist propanolol induced a suppression of the odour-related patterns of Fos accumulation similar to the one caused by the surgical deafferentation of the olfactory bulb. Propranolol 31-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 8793085-9 1996 The muscarinic antagonist scopolamine did not alter c-fos expression in the odour-selective area but increased significantly Fos labelling in the other bulbar aspects. Scopolamine 26-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-128 8813576-0 1996 Contrasting effects of SM-9018, a potential atypical antipsychotic, and haloperidol on c-fos mRNA expression in the rat striatum. perospirone 23-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 8813576-0 1996 Contrasting effects of SM-9018, a potential atypical antipsychotic, and haloperidol on c-fos mRNA expression in the rat striatum. Haloperidol 72-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 8813576-2 1996 Northern blot analysis was performed to compare the effects of SM-9018 and of haloperidol on the striatal c-fos mRNA expression in rats. Haloperidol 78-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 8813576-4 1996 markedly increased the striatal c-fos mRNA levels (about eight-fold at 30 mg/kg), the increase being abolished by lesioning of dopamine neurons with 6-hydroxydopamine. Dopamine 127-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 8813576-4 1996 markedly increased the striatal c-fos mRNA levels (about eight-fold at 30 mg/kg), the increase being abolished by lesioning of dopamine neurons with 6-hydroxydopamine. Oxidopamine 149-166 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 8723713-5 1996 All DA agonists significantly increased c-fos levels after apomorphine injection. Apomorphine 59-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 8723713-1 1996 Striatal c-fos levels and stereotyped behavior have been evaluated in chronically haloperidol-treated rats which received subsequent subacute dopamine (DA) agonist treatment to investigate the possible relationship between striatal c-fos and behavioral supersensitivity. Haloperidol 82-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 232-237 8761989-0 1996 Antisense oligodeoxynucleotide attenuates in vivo expression of c-fos in the paraventricular hypothalamic nucleus of the rat brain. Oligodeoxyribonucleotides 10-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 8761989-3 1996 Our results indicate that c-fos antisense oligodeoxynucleotide treatment decreases the density of Fos immuno-labeled nuclei in the PVN following IL-1 beta administration. Oligodeoxyribonucleotides 42-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 8761989-3 1996 Our results indicate that c-fos antisense oligodeoxynucleotide treatment decreases the density of Fos immuno-labeled nuclei in the PVN following IL-1 beta administration. Oligodeoxyribonucleotides 42-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 8782869-1 1996 Expression of the inducible transcription factor c-Fos has been examined in the lumbar spinal cord following noxious chemical stimulation (injection of 2% formalin) of the ankles or the ventral skin of the hindpaws of either normal rats, or monoarthritic rats during the chronic phase of the disease. Formaldehyde 155-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 8782869-5 1996 One hour after formalin stimulation of the ankle or hindpaw skin of normal rats expression of c-Fos was observed throughout the ipsilateral, but not contralateral dorsal horn. Formaldehyde 15-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 8782869-6 1996 Formalin stimulation of the inflamed ankle in four-week arthritic rats induced a 3-to-6 fold increase in c-Fos expression in the ipsilateral dorsal horn compared to formalin stimulation of the ankle in normal rats. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 8813576-7 1996 ), dose-dependently attenuated the haloperiodol-induced c-fos expression, but the putative 5-HT1A receptor antagonist, NAN-190 (1-(2-methoxyphenyl)-4-(4-(2-phethalimmido)butyl)piperazine HBr; 1-10 mg/kg, i.p. haloperiodol 35-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 8928931-0 1996 Role of NMDA and AMPA glutamatergic transmission in spinal c-fos expression after urinary tract irritation. N-Methylaspartate 8-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 8928931-4 1996 Systemic administration of LY-215490 produced a dose-dependent decrease in the number of Fos-positive cells after LUT irritation in the DCM and SPN areas, whereas in the DH only the highest dose (10 mg/kg) of LY-215490 decreased the number of Fos-positive cells. tezampanel 27-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 8928931-4 1996 Systemic administration of LY-215490 produced a dose-dependent decrease in the number of Fos-positive cells after LUT irritation in the DCM and SPN areas, whereas in the DH only the highest dose (10 mg/kg) of LY-215490 decreased the number of Fos-positive cells. tezampanel 27-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 243-246 8928931-4 1996 Systemic administration of LY-215490 produced a dose-dependent decrease in the number of Fos-positive cells after LUT irritation in the DCM and SPN areas, whereas in the DH only the highest dose (10 mg/kg) of LY-215490 decreased the number of Fos-positive cells. Lysine 27-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 8928931-4 1996 Systemic administration of LY-215490 produced a dose-dependent decrease in the number of Fos-positive cells after LUT irritation in the DCM and SPN areas, whereas in the DH only the highest dose (10 mg/kg) of LY-215490 decreased the number of Fos-positive cells. Lysine 27-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 243-246 8928931-6 1996 However, a combined administration of low doses of MK-801 and LY-215490 significantly decreased the number of Fos-positive cells in all regions of the spinal cord. Dizocilpine Maleate 51-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 8928931-6 1996 However, a combined administration of low doses of MK-801 and LY-215490 significantly decreased the number of Fos-positive cells in all regions of the spinal cord. Lysine 62-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 8626609-4 1996 Induction of c-fos by ouabain was prevented when either extracellular or intracellular Ca2+ was lowered and was attenuated by pretreatment of myocytes with a phorbol ester under conditions known to down-regulate protein kinase C. Exposure to ouabain for 24-48 h also increased total transcriptional activity and protein content of myocytes. Ouabain 22-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 8782869-8 1996 Formalin stimulation of the contralateral ankle in monoarthritic rats (i.e. the non-inflamed ankle) induced an ipsilateral expression of c-Fos which was similar to that observed after stimulation of the arthritic ankle. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 8782869-10 1996 Finally, formalin stimulation of the hindpaw skin (which was not inflamed) of the arthritic limb induced the same number of c-Fos labelled cells in the superficial laminae as did formalin stimulation of the skin of normal rats; but in the deep laminae there was a 1.6-fold increase in the number of labelled cells. Formaldehyde 9-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 8782873-0 1996 Amphetamine induces Fos-like immunoreactivity in the striatum of primates. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 8736437-7 1996 By lowering circulating levels of the PRL with bromocryptine-or PRL antiserum-treatment, we noticed a decrease in the number of (beta-endorphin + Fos)-ir neurons compared to non-injected freely nursing lactating females. Bromocriptine 47-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-149 8627519-2 1996 Saline-treated rats processed in the forced swim test exhibited marked increases in Fos-LI in limbic cortical regions, lateral septum, medial amygdala and paraventricular nucleus of the hypothalamus (PVN). Sodium Chloride 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 8627519-3 1996 Uptake of 2-DG was increased by swim stress in some of the same brain regions where Fos-LI was induced, with the notable exception of a lack of a change in the PVN. Deoxyglucose 10-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 8627519-5 1996 Chronic treatment with imipramine and desipramine alone induced Fos-LI in the central nucleus of the amygdala and the dorsolateral bed nucleus of the stria terminalis. Imipramine 23-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 8627519-5 1996 Chronic treatment with imipramine and desipramine alone induced Fos-LI in the central nucleus of the amygdala and the dorsolateral bed nucleus of the stria terminalis. Desipramine 38-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 8627519-6 1996 After tranylcypromine treatment, Fos-LI was induced in many brain regions including limbic cortex, amygdala and paraventricular nucleus of the hypothalamus. Tranylcypromine 6-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 8627519-8 1996 Chronic administration of imipramine, desipramine and nisoxetine antagonized the swim induced expression of Fos-LI in the PVN and in limbic cortical regions, including the medial prefrontal ventrolateral orbital and cingulate cortices. Imipramine 26-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 8627519-8 1996 Chronic administration of imipramine, desipramine and nisoxetine antagonized the swim induced expression of Fos-LI in the PVN and in limbic cortical regions, including the medial prefrontal ventrolateral orbital and cingulate cortices. Desipramine 38-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 8627519-8 1996 Chronic administration of imipramine, desipramine and nisoxetine antagonized the swim induced expression of Fos-LI in the PVN and in limbic cortical regions, including the medial prefrontal ventrolateral orbital and cingulate cortices. nisoxetine 54-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 8627519-10 1996 Thus, only antidepressant drugs that affect norepinephrine uptake (i.e., imipramine, desipramine and nisoxetine) antagonized swim stress-induced Fos-LI. Norepinephrine 44-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-148 8627519-10 1996 Thus, only antidepressant drugs that affect norepinephrine uptake (i.e., imipramine, desipramine and nisoxetine) antagonized swim stress-induced Fos-LI. Imipramine 73-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-148 8627519-10 1996 Thus, only antidepressant drugs that affect norepinephrine uptake (i.e., imipramine, desipramine and nisoxetine) antagonized swim stress-induced Fos-LI. Desipramine 85-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-148 8627519-10 1996 Thus, only antidepressant drugs that affect norepinephrine uptake (i.e., imipramine, desipramine and nisoxetine) antagonized swim stress-induced Fos-LI. nisoxetine 101-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-148 8627519-12 1996 Acute administration of propranolol, which blocks beta-adrenergic receptors, reduced the number of cells staining for Fos-LI in limbic cortical regions, resembling the effects produced by chronic imipramine, desipramine and nisoxetine. Propranolol 24-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 8627519-12 1996 Acute administration of propranolol, which blocks beta-adrenergic receptors, reduced the number of cells staining for Fos-LI in limbic cortical regions, resembling the effects produced by chronic imipramine, desipramine and nisoxetine. Imipramine 196-206 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 8728564-7 1996 In diazepam- and nicotine-treated rats Fos IS was increased in PVN and SON as well as in MT and i.p.. Diazepam 3-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 8728564-7 1996 In diazepam- and nicotine-treated rats Fos IS was increased in PVN and SON as well as in MT and i.p.. Nicotine 17-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 8728564-9 1996 of diazepam and nicotine-treated rats Fos IS was similar to that induced by nicotine alone, and in PVN and SON of these rats Fos IS in ACe. Diazepam 3-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 8728564-9 1996 of diazepam and nicotine-treated rats Fos IS was similar to that induced by nicotine alone, and in PVN and SON of these rats Fos IS in ACe. Nicotine 16-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 8728564-10 1996 Taken together, diazepam induced Fos IS in all stress-related areas studied (PVN, SON, ACe), but not in central visual structures, where nicotine induces Fos IS (MT, i.p.). Diazepam 16-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 8728564-10 1996 Taken together, diazepam induced Fos IS in all stress-related areas studied (PVN, SON, ACe), but not in central visual structures, where nicotine induces Fos IS (MT, i.p.). Diazepam 16-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-157 8728564-10 1996 Taken together, diazepam induced Fos IS in all stress-related areas studied (PVN, SON, ACe), but not in central visual structures, where nicotine induces Fos IS (MT, i.p.). Nicotine 137-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-157 8795081-11 1996 These sites were previously found to show strong Fos-IR after administration of either cholecystokinin (CCK) or dexfenfluramine (DF), but with different subregional distribution. Dexfenfluramine 112-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 8782874-3 1996 Hypertonic saline injection with and without anesthesia induced Fos-ir in 66% and 77% of SON neurons, respectively, whereas isotonic saline with anesthesia and anesthesia alone resulted in 15% and 13%, respectively, of cells showing Fos-ir. Sodium Chloride 11-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 8782874-3 1996 Hypertonic saline injection with and without anesthesia induced Fos-ir in 66% and 77% of SON neurons, respectively, whereas isotonic saline with anesthesia and anesthesia alone resulted in 15% and 13%, respectively, of cells showing Fos-ir. Sodium Chloride 11-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 233-236 8737671-4 1996 However, the expression of c-fos mRNA was comparable in neocortex, dorsal hippocampus and medial geniculate body between control rats and PTU-treated, seizure-induced rats. Propylthiouracil 138-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 8730712-0 1996 P-chloroamphetamine induces c-fos in rat brain: a study of serotonin2A/2C receptor function. p-Chloroamphetamine 0-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 8730712-4 1996 Administration of p-chloroamphetamine or the serotonin2A/2C receptor agonist 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane to rats resulted in similar patterns of Fos-like immunoreactivity in some, but not all, forebrain areas. p-Chloroamphetamine 18-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 8730712-4 1996 Administration of p-chloroamphetamine or the serotonin2A/2C receptor agonist 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane to rats resulted in similar patterns of Fos-like immunoreactivity in some, but not all, forebrain areas. 4-iodo-2,5-dimethoxyphenylisopropylamine 77-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 8730712-8 1996 Prior treatment of animals with p-chloroamphetamine two weeks before a second challenge with the same drug, or with the serotonin2A/2C receptor antagonist ritanserin 30 min before p-chloroamphetamine challenge, resulted in an attenuation of p-chloroamphetamine-induced Fos-like immunoreactivity in the olfactory tubercle, the islands of Calleja and the caudate-putamen. p-Chloroamphetamine 32-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 269-272 8730712-8 1996 Prior treatment of animals with p-chloroamphetamine two weeks before a second challenge with the same drug, or with the serotonin2A/2C receptor antagonist ritanserin 30 min before p-chloroamphetamine challenge, resulted in an attenuation of p-chloroamphetamine-induced Fos-like immunoreactivity in the olfactory tubercle, the islands of Calleja and the caudate-putamen. Ritanserin 155-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 269-272 8730712-8 1996 Prior treatment of animals with p-chloroamphetamine two weeks before a second challenge with the same drug, or with the serotonin2A/2C receptor antagonist ritanserin 30 min before p-chloroamphetamine challenge, resulted in an attenuation of p-chloroamphetamine-induced Fos-like immunoreactivity in the olfactory tubercle, the islands of Calleja and the caudate-putamen. chloroamphetamine 34-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 269-272 8730713-6 1996 Our results show that burst stimulation of the medial forebrain bundle, which increase release of dopamine in target areas, increases the basal Fos-like immunoreactivity in the stimulated hemisphere, while regular stimulation does not affect expression of this protein. Dopamine 98-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-147 8730718-5 1996 Systemic administration of methamphetamine (1-5 mg/kg) inhibited light (300 lux, 1h)-induced Fos expression in the suprachiasmatic nucleus; methamphetamine at a dose of 5 mg/kg, i.p. Methamphetamine 27-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 8730718-5 1996 Systemic administration of methamphetamine (1-5 mg/kg) inhibited light (300 lux, 1h)-induced Fos expression in the suprachiasmatic nucleus; methamphetamine at a dose of 5 mg/kg, i.p. Hydrogen 81-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 8730720-6 1996 Naltrexone-precipitated abstinence resulted in an increase in Fos expression at all levels of the spinal cord; the greatest increase and densest staining was in laminae I through VI. Naltrexone 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 8730721-7 1996 Further, the magnitudes of the bimodal peaks of Fos produced by 52 degrees C thermal probe and mustard oil stimuli were different quantitatively. mustard oil 95-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 8730721-8 1996 Mustard oil caused a greater Fos response at the subnucleus interpolaris/caudalis transition than 52 degrees C thermal probe stimulation, whereas the opposite was true at the subnucleus caudalis/cervical cord transition. mustard oil 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 8737422-2 1996 Since the stomach is continually exposed to injurious chemicals, the present study examined whether application of acid (0.15 M HCl) and formalin (5%) to the gastric mucosa or serosal surface of the stomach stimulates c-fos transcription in the caudal thoracic spinal cord of anaesthetized rats. Formaldehyde 137-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 218-223 8737422-5 1996 Application of acid to the serosal surface of the stomach was also unable to stimulate c-fos transcription, whereas serosal application of formalin led to substantial expression of c-fos messenger RNA in the superficial but also deeper laminae of the spinal dorsal horn when examined 45 min, but not 15 or 120 min, post-stimulation. Formaldehyde 139-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 181-186 8737422-6 1996 The highest expression of c-fos messenger RNA was seen when formalin was injected subcutaneously into one hindpaw and c-fos transcription was examined in the lumbar spinal cord. Formaldehyde 60-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 8626609-4 1996 Induction of c-fos by ouabain was prevented when either extracellular or intracellular Ca2+ was lowered and was attenuated by pretreatment of myocytes with a phorbol ester under conditions known to down-regulate protein kinase C. Exposure to ouabain for 24-48 h also increased total transcriptional activity and protein content of myocytes. Phorbol Esters 158-171 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 8626609-4 1996 Induction of c-fos by ouabain was prevented when either extracellular or intracellular Ca2+ was lowered and was attenuated by pretreatment of myocytes with a phorbol ester under conditions known to down-regulate protein kinase C. Exposure to ouabain for 24-48 h also increased total transcriptional activity and protein content of myocytes. Ouabain 242-249 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 8725293-11 1996 Significantly more neurons were FOS-labeled in PBdl, PBsl, and PBcl on the side contralateral to the hemisection than on the ipsilateral side. pbdl 47-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 8725293-11 1996 Significantly more neurons were FOS-labeled in PBdl, PBsl, and PBcl on the side contralateral to the hemisection than on the ipsilateral side. pbsl 53-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 8725293-11 1996 Significantly more neurons were FOS-labeled in PBdl, PBsl, and PBcl on the side contralateral to the hemisection than on the ipsilateral side. pbcl 63-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 8619843-1 1996 This study shows that induction of heme oxygenase-1 (HO-1) gene expression by a glutathione (GSH) depletor, phorone, is inhibited by cycloheximide pretreatment and involves changes in c-jun, not c-fos, mRNA. Glutathione 80-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-200 8728564-0 1996 Expression of Fos protein in various rat brain areas following acute nicotine and diazepam. Nicotine 69-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 8728564-0 1996 Expression of Fos protein in various rat brain areas following acute nicotine and diazepam. Diazepam 82-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 8728564-1 1996 We studied the effects of an acute dose of (-)-nicotine (1 mg/kg) on Fos-like immunostaining (IS) in rat brain areas. Nicotine 43-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 8728564-2 1996 Nicotine increased Fos IS significantly in the medial terminal nucleus of accessory optic tract (MT), and tended to increase it in the interpeduncular nucleus (i.p. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 8728564-4 1996 Previously nicotine was reported to increase Fos IS also in another stress-related area, the central nucleus of amygdala (ACe). Nicotine 11-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 8728564-6 1996 Diazepam alone increased Fos IS in PVN and in SON as well as in ACe. Diazepam 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 8738241-0 1996 Amphetamine-induced Fos expression in globus pallidus is altered by frontal cortex injury. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 8738241-3 1996 In the ipsilateral dorsal globus pallidus of rats demonstrating neglect of contralateral stimuli (sacrificed 5 days postinjury), the numbers of amphetamine-induced Fos-positive nuclei were reduced 37% compared to intact hemisphere values. Amphetamine 144-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-167 8622865-1 1996 c-Fos is phosphorylated by MAP kinase and the 90 kDa-ribosomal S6 kinase (RSK) in vitro at serines 362 and 374 (rat) which we demonstrate are major in vivo phosphorylation sites in early G1. Serine 91-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8622865-2 1996 We have constructed c-Fos mutants with these serines changed to aspartic acid residues (FosD) to mimic phosphorylation or to alanine residues (FosA) to prevent phosphorylation. Serine 45-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 8622865-2 1996 We have constructed c-Fos mutants with these serines changed to aspartic acid residues (FosD) to mimic phosphorylation or to alanine residues (FosA) to prevent phosphorylation. Aspartic Acid 64-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 8637502-2 1996 In addition, this enzyme has been shown to function as a redox factor facilitating the DNA binding capability of Jun-Jun homodimers and Fos-Jun heterodimers by altering their redox state and to be involved in calcium mediated transcriptional repression of the parathyroid hormone gene. Calcium 209-216 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-139 8859908-1 1996 Acute seizures and status epilepticus induced by pilocarpine lead to the expression of Fos-like immunoreactivity in several specific brain areas in a manner similar to that of other models of limbic seizures. Pilocarpine 49-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 8739631-2 1996 In order to investigate central pathways mediating reflex changes in cardiovascular activity, immunohistochemical localization of cells expressing the immediate-early gene, c-fos, was used to identify central nervous responding to noxious electrical stimulation of mandibular, incisor tooth dentin or chemical (capsaicin) stimulation of tooth pulp in the anesthetized rat. Capsaicin 311-320 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 173-178 8739641-2 1996 ), a selective cyclooxygenase-2 inhibitor without gastro-intestinal side-effects, dose-dependently reduced carrageenan evoked spinal c-Fos expression (16 +/- 4%, 32 +/- 3% and 56 +/- 5% reduction, respectively) at 3 h after intraplantar carrageenan. Carrageenan 107-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 8619843-1 1996 This study shows that induction of heme oxygenase-1 (HO-1) gene expression by a glutathione (GSH) depletor, phorone, is inhibited by cycloheximide pretreatment and involves changes in c-jun, not c-fos, mRNA. Glutathione 93-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-200 8619843-1 1996 This study shows that induction of heme oxygenase-1 (HO-1) gene expression by a glutathione (GSH) depletor, phorone, is inhibited by cycloheximide pretreatment and involves changes in c-jun, not c-fos, mRNA. phorone 108-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-200 8619843-1 1996 This study shows that induction of heme oxygenase-1 (HO-1) gene expression by a glutathione (GSH) depletor, phorone, is inhibited by cycloheximide pretreatment and involves changes in c-jun, not c-fos, mRNA. Cycloheximide 133-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-200 8635209-1 1996 We have previously shown that extracellular ATP, like norepinephrine (NE) and many other hypertrophy-inducing agents, increases expression of the immediate-early genes c-fos and junB in cultured neonatal cardiac myocytes but that the intracellular signaling pathways activated by ATP and responsible for these changes differ from those stimulated by NE. Adenosine Triphosphate 44-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-173 8738141-0 1996 Fenfluramine-induced activation of the immediate-early gene c-fos in the striatum: possible interaction between serotonin and dopamine. Fenfluramine 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 8738141-0 1996 Fenfluramine-induced activation of the immediate-early gene c-fos in the striatum: possible interaction between serotonin and dopamine. Serotonin 112-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 8738141-0 1996 Fenfluramine-induced activation of the immediate-early gene c-fos in the striatum: possible interaction between serotonin and dopamine. Dopamine 126-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 8738141-13 1996 Our results suggest that the release of 5-HT by fenfluramine induced Fos-LI expression predominantly in a striatal region related to associative functions and, that this c-fos response may be under the control of both 5-HT and DA. Fenfluramine 48-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-175 8738141-14 1996 Moreover, the mechanism by which fenfluramine induces c-fos expression in the striatum differs from other brain regions. Fenfluramine 33-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 8625905-9 1996 To determine if addition of genistein to FRTL-5 cells resulted in a general inhibition of Bu2cAMP-induced responses, we examined its effect on the Bu2cAMP-induced increase in c-fos mRNA levels. Genistein 28-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 175-180 8625905-9 1996 To determine if addition of genistein to FRTL-5 cells resulted in a general inhibition of Bu2cAMP-induced responses, we examined its effect on the Bu2cAMP-induced increase in c-fos mRNA levels. bu2camp 147-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 175-180 8625905-10 1996 Bu2cAMP-induced c-fos mRNA levels were not affected by the treatment of cells with genistein (100 microM). bu2camp 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 8635209-1 1996 We have previously shown that extracellular ATP, like norepinephrine (NE) and many other hypertrophy-inducing agents, increases expression of the immediate-early genes c-fos and junB in cultured neonatal cardiac myocytes but that the intracellular signaling pathways activated by ATP and responsible for these changes differ from those stimulated by NE. Norepinephrine 54-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-173 8740208-2 1996 The possibility that attenuation of nociceptive messages from the cornea by diltiazem reduced Fos-like immunoreactivity of spinal trigeminal neurons was also examined. Diltiazem 76-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 8609891-0 1996 Induction of chronic Fos-related antigens in rat brain by chronic morphine administration. Morphine 66-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 8609891-1 1996 Previous studies have shown that repeated exposure to cocaine or to several other stimuli induces novel 35-37 kDa Fos-related antigens (chronic Fras) in specific brain regions. Cocaine 54-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 8684609-15 1996 An endothelin-A receptor antagonist, BQ-123, abolished c-fos expression in all structures in the forebrain and brainstem following endothelin-1 infusions. cyclo(Trp-Asp-Pro-Val-Leu) 37-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 8826484-0 1996 Inhibition of noxious stimulus-evoked pain behaviors and neuronal fos-like immunoreactivity in the spinal cord of the rat by supraspinal morphine. Morphine 137-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 8826484-1 1996 In previous studies, we reported that supraspinally administered DAMGO, a mu-opioid agonist, produces a dose-related, naloxone-reversible inhibition of formalin-evoked pain behaviors and spinal cord Fos-like immunoreactivity (FLI) in the rat spinal cord. Naloxone 118-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-202 8826484-8 1996 morphine tested (0.70 nmol) produced a significant reduction of FLI in the superficial laminae without producing behavioral antinociception, which is consistent with our hypothesis that noxious stimulus-evoked Fos expression in the superficial laminae is a poor predictor of the magnitude of pain behavior. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 210-213 8927504-0 1996 Effect of sinus denervation and vagotomy on c-fos expression in the nucleus tractus solitarius after exposure to CO2. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 113-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 8927504-3 1996 After exposure to CO2 or air the rats were anesthetized, perfused with 4% paraformaldehyde and the brains processed for immunohistochemical staining for c-fos protein using the PAP (i.e. peroxidase anti-peroxidase) technique. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 18-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-158 8733883-12 1996 The differential induction of fos and jun family genes suggests an important role of their gene products on the regulation of thyroid cell function by TSH. Thyrotropin 151-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 9389166-4 1996 When both NRO and dPAG were stimulated, Fos- like immunoreactive (FLI) cells in the ventral periaqueductal grey (vPAG) and paragigantocellularis lateralis (PGL) were increased as compared with cases in which the stimulation was only delivered to dPAG, i.e., reaching a FLI cell count value of 66.5 +/- 8.3 and 10.8 +/- 1.5 respectively. dpag 18-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 9389166-4 1996 When both NRO and dPAG were stimulated, Fos- like immunoreactive (FLI) cells in the ventral periaqueductal grey (vPAG) and paragigantocellularis lateralis (PGL) were increased as compared with cases in which the stimulation was only delivered to dPAG, i.e., reaching a FLI cell count value of 66.5 +/- 8.3 and 10.8 +/- 1.5 respectively. dpag 246-250 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 9592236-2 1996 The results were: (a) 2Hz and 100Hz EA induced differential Fos expression in different brain areas; (b) EA of both frequencies accelerated PPE gene transcription, but 2Hz EA was more effective than 100Hz EA; (c) PPD expression was accelerated by 100Hz EA, but not by 2Hz EA; (d) the blockade of Fos/Jun expression by c-fos/c-jun antisense ODNs prevented EA from accelerating PPD but not PPE mRNA expression. (2z)-3-(3'-Nitrobiphenyl-3-Yl)prop-2-Enoic Acid 22-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 8724971-3 1996 Intravenous infusions of hypertonic saline resulted in dense Fos-like immunoreactivity in several forebrain (paraventricular nucleus of the hypothalamus (PVH), supraoptic nucleus (SON), median preoptic nucleus (MnPO), medial preoptic nucleus, organum vasculosum of the laminae terminalis and (SFO) and brainstem (nucleus of the solitary tract, ventrolateral medulla, and parabrachial nucleus) structures. Sodium Chloride 36-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 8724971-4 1996 Intravenous infusions of the hypertonic saline solution into animals with lesions of either the SFO, the AV3V or both resulted in a decreased number of Fos-like immunoreactive neurons in the MnPO, PVH and SON. Sodium Chloride 29-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-155 8724971-5 1996 In addition, the number of Fos-labeled neurons in the SON after lesions of both the SFO and the AV3V was significantly greater than that observed in isotonic saline infused controls. Sodium Chloride 158-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 8625290-8 1996 This pattern of gene expression is consistent with that induced by other cytotoxic carcinogens and suggest that alteration of the myc and fos genes could be involved in the regenerative cell proliferation that ultimately could play a role in chloroform-induced tumors. Chloroform 242-252 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-141 8814891-0 1996 Fos-like immunoreactivity in the mamillary body and thalamus following injections of muscimol into the ventral tegmental nucleus of Gudden in the rat. Muscimol 85-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 8708002-8 1996 Like the competitive antagonists, intrastriatal infusion of the non-competitive NMDA antagonist MK-801 partially decreased c-fos, but not zif268, mRNA in the area around the microdialysis probe. N-Methylaspartate 80-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 8708002-8 1996 Like the competitive antagonists, intrastriatal infusion of the non-competitive NMDA antagonist MK-801 partially decreased c-fos, but not zif268, mRNA in the area around the microdialysis probe. Dizocilpine Maleate 96-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 8708002-0 1996 D1 dopamine receptor-mediated induction of zif268 and c-fos in the dopamine-depleted striatum: differential regulation and independence from NMDA receptors. Dopamine 3-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 8708002-5 1996 Intrastriatal infusion of the competitive NMDA receptor antagonist (+/-)-3-(2-carboxypiperazin-4-yl)-propyl -1-phosphonic acid caused a dose-dependent attenuation of SKF 38393-induced rotation and partially decreased c-fos mRNA expression. 3-(2-carboxypiperazin-4-yl)propyl-1-phosphonic acid 67-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 217-222 8814891-2 1996 The current study examined Fos-like immunoreactivity in the mamillary body and the anterior thalamus following unilateral injections of the inhibitory GABA-A agonist muscimol into the ventral tegmental nucleus of Gudden (VTN). gamma-Aminobutyric Acid 151-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 8708002-9 1996 However, unlike competitive antagonists, local infusion of 1 mM MK-801 potentiated D1-mediated increases in c-fos and zif268 mRNAs in lateral striatum. Dizocilpine Maleate 64-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 8814891-2 1996 The current study examined Fos-like immunoreactivity in the mamillary body and the anterior thalamus following unilateral injections of the inhibitory GABA-A agonist muscimol into the ventral tegmental nucleus of Gudden (VTN). Muscimol 166-174 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 8814899-8 1996 In sum, the present findings indicate that AMPH-induced Fos expression is sexually dimorphic and modulated by gonadal hormones in lateral regions of the rat dorsal striatum. Amphetamine 43-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 8710170-5 1996 Both in animals with intrathalamic and in animals with intracortical grafts, systemic administration of a low dose of apomorphine (0.25 mg/kg) induced intense fos expression in striatum-like patches not innervated by dopaminergic fibres. Apomorphine 118-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-162 8907361-1 1996 Immunohistochemical detection of the protein product (Fos) of the c-fos immediate early gene was used to study neuronal activation in the rostral pons and midbrain of halothane-anesthetised rats following noxious deep somatic or noxious visceral stimulation. Halothane 167-176 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 8907361-1 1996 Immunohistochemical detection of the protein product (Fos) of the c-fos immediate early gene was used to study neuronal activation in the rostral pons and midbrain of halothane-anesthetised rats following noxious deep somatic or noxious visceral stimulation. Halothane 167-176 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 8907361-2 1996 In animals exposed only to halothane anesthesia, Fos-like immunoreactive (IR) neurons were located in the midbrain periaqueductal gray matter, tectum, and parabrachial nucleus. Halothane 27-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 8907361-3 1996 Following noxious stimulation of hindlimb muscle, knee joint, vagal cardiopulmonary, or peritoneal nociceptors, there was, compared to halothane-only animals, a significant increase in the numbers of Fos-like (IR) cells in the caudal ventrolateral periaqueductal gray and the intermediate gray lamina of the superior colliculus. Halothane 135-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 200-203 8709975-0 1996 Regulation of c-fos mRNA expression in Sertoli cells by cyclic AMP, calcium, and protein kinase C mediated pathways. Cyclic AMP 56-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 8709975-3 1996 Although the action of FSH on Sertoli cells is considered to be mediated by cAMP, dibutyryl cAMP (dbcAMP), a potent membrane permeable analog of cAMP, induced much less c-fos mRNA expression than FSH ( < 50%) suggesting that additional cAMP-independent mechanisms may mediate the effect of FSH on c-fos. Bucladesine 82-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-174 8709975-3 1996 Although the action of FSH on Sertoli cells is considered to be mediated by cAMP, dibutyryl cAMP (dbcAMP), a potent membrane permeable analog of cAMP, induced much less c-fos mRNA expression than FSH ( < 50%) suggesting that additional cAMP-independent mechanisms may mediate the effect of FSH on c-fos. Bucladesine 82-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 300-305 8709975-3 1996 Although the action of FSH on Sertoli cells is considered to be mediated by cAMP, dibutyryl cAMP (dbcAMP), a potent membrane permeable analog of cAMP, induced much less c-fos mRNA expression than FSH ( < 50%) suggesting that additional cAMP-independent mechanisms may mediate the effect of FSH on c-fos. Bucladesine 98-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-174 8709975-3 1996 Although the action of FSH on Sertoli cells is considered to be mediated by cAMP, dibutyryl cAMP (dbcAMP), a potent membrane permeable analog of cAMP, induced much less c-fos mRNA expression than FSH ( < 50%) suggesting that additional cAMP-independent mechanisms may mediate the effect of FSH on c-fos. Bucladesine 98-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 300-305 8709975-3 1996 Although the action of FSH on Sertoli cells is considered to be mediated by cAMP, dibutyryl cAMP (dbcAMP), a potent membrane permeable analog of cAMP, induced much less c-fos mRNA expression than FSH ( < 50%) suggesting that additional cAMP-independent mechanisms may mediate the effect of FSH on c-fos. Cyclic AMP 92-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-174 8709975-3 1996 Although the action of FSH on Sertoli cells is considered to be mediated by cAMP, dibutyryl cAMP (dbcAMP), a potent membrane permeable analog of cAMP, induced much less c-fos mRNA expression than FSH ( < 50%) suggesting that additional cAMP-independent mechanisms may mediate the effect of FSH on c-fos. Cyclic AMP 92-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 300-305 8709975-3 1996 Although the action of FSH on Sertoli cells is considered to be mediated by cAMP, dibutyryl cAMP (dbcAMP), a potent membrane permeable analog of cAMP, induced much less c-fos mRNA expression than FSH ( < 50%) suggesting that additional cAMP-independent mechanisms may mediate the effect of FSH on c-fos. Cyclic AMP 92-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-174 8709975-3 1996 Although the action of FSH on Sertoli cells is considered to be mediated by cAMP, dibutyryl cAMP (dbcAMP), a potent membrane permeable analog of cAMP, induced much less c-fos mRNA expression than FSH ( < 50%) suggesting that additional cAMP-independent mechanisms may mediate the effect of FSH on c-fos. Cyclic AMP 92-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 300-305 8709975-5 1996 Ionomycin, which increases intracellular free calcium concentration, induced c-fos expression significantly. Ionomycin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 8709975-5 1996 Ionomycin, which increases intracellular free calcium concentration, induced c-fos expression significantly. Calcium 46-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 8709975-6 1996 These data demonstrate that Sertoli cell c-fos mRNA expression is under multifactorial regulation by cAMP, calcium, and PKC. Cyclic AMP 101-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 8709975-6 1996 These data demonstrate that Sertoli cell c-fos mRNA expression is under multifactorial regulation by cAMP, calcium, and PKC. Calcium 107-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 8632353-0 1996 Differential contribution of descending controls to the antinociceptive actions of kappa and mu opioids: an analysis of formalin-evoked C-fos expression. Formaldehyde 120-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 8882592-13 1996 In the parvocellular division of the PVN, the large majority of CRF-ir perikarya displayed a positive signal for the mRNA encoding c-fos, indicating a profound CRFergic activation within this neuroendocrine nucleus after dexfenfluramine administration. Dexfenfluramine 221-236 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-136 8603591-7 1996 The present study, however, indicates that IL-1 induced expression of Fos and Jun does not seem to participate in the regulation of iNOS and mRNA expression, because: 1) cycloheximide (1 microM) completely inhibited Fos expression but had no inhibitory effect on iNOS mRNA levels; and 2) tyrosine kinase inhibitors genistein and herbimycin A completely inhibited IL-1 induced iNOS expression but did not block c-fos and c-jun expression. Cycloheximide 170-183 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 216-219 8698450-3 1996 In WKY, 474 +/- 56 (n=6) Fos-positive neurons were identified in the rostral ventral medulla after nitroprusside infusion, a fivefold increase from controls; 50% of the tyrosine hydroxylase-containing neurons in the rostral ventral medulla were activated by this hypotension. Nitroprusside 99-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 8698450-4 1996 Sympathetic preganglionic neurons, mainly sympathoadrenal neurons, were Fos positive after nitroprusside, but Fos-positive sympathetic preganglionic neurons were not observed in control WKY. Nitroprusside 91-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 8783322-9 1996 respectively), which had negligible effects when administered separately, greatly reduced both the total number of carrageenan-evoked c-Fos-LI neurones (61 +/- 5% reduction as compared to control value) and the peripheral oedema (80 +/- 8% and 60 +/- 5% reduction for ankle and paw oedema, respectively). Carrageenan 115-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-139 8783322-10 1996 The attenuation by co-administered dexamethasone and diclofenac, of both c-Fos expression and the peripheral oedema, was significantly greater than the effect of dexamethasone alone (P < 0.001 for both) and diclofenac alone (P < 0.001 for both). Dexamethasone 35-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 8783322-10 1996 The attenuation by co-administered dexamethasone and diclofenac, of both c-Fos expression and the peripheral oedema, was significantly greater than the effect of dexamethasone alone (P < 0.001 for both) and diclofenac alone (P < 0.001 for both). Diclofenac 53-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 8783322-11 1996 Our study illustrates enhanced attenuating effects of co-administered dexamethasone and diclofenac on both inflammatory oedema and the associated spinal expression of c-Fos, an indicator of nociceptive transmission at the spinal level. Dexamethasone 70-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 167-172 8783322-11 1996 Our study illustrates enhanced attenuating effects of co-administered dexamethasone and diclofenac on both inflammatory oedema and the associated spinal expression of c-Fos, an indicator of nociceptive transmission at the spinal level. Diclofenac 88-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 167-172 8963664-0 1996 Roles of dopamine D1 receptors in delta 9-tetrahydrocannabinol-induced expression of Fos protein in the rat brain. Dronabinol 34-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 8963664-1 1996 We examined whether administration of delta 9-tetrahydrocannabinol (THC) induces the expression of Fos protein or not in the rat brain. Dronabinol 38-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 8963664-1 1996 We examined whether administration of delta 9-tetrahydrocannabinol (THC) induces the expression of Fos protein or not in the rat brain. Dronabinol 68-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 8963664-2 1996 A single administration of 3.2 and 10 mg/kg THC produced a dose-dependent and significant increase in Fos-immunoreactive cells in the striatum, particularly in its dorsomedial portions. Dronabinol 44-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 8963664-3 1996 The peak increase was reached 2 h after THC treatment and was absent at 8 h. Fos induction was also observed in the nucleus accumbens after administration of 10 mg/kg THC. Dronabinol 40-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-80 8963664-3 1996 The peak increase was reached 2 h after THC treatment and was absent at 8 h. Fos induction was also observed in the nucleus accumbens after administration of 10 mg/kg THC. Dronabinol 167-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-80 8963664-5 1996 SCH-23390, a selective dopamine D1 receptor antagonist, at 0.32 mg/kg produced a significant block of the effects of THC on Fos expression in the striatum and the nucleus accumbens. SCH 23390 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-127 8963664-5 1996 SCH-23390, a selective dopamine D1 receptor antagonist, at 0.32 mg/kg produced a significant block of the effects of THC on Fos expression in the striatum and the nucleus accumbens. Dronabinol 117-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-127 8963664-7 1996 These findings indicate that THC induces the expression of Fos protein and that this expression is mediated at least by dopamine D1 receptors. Dronabinol 29-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 8626753-6 1996 Additionally, H2O2 increased the mRNA expression of MAPK-dependent genes c-jun, c-fos, and MAPK phosphatase-1. Hydrogen Peroxide 14-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 8652063-3 1996 Peripherally induced hyposmia by ZnSo4 reduced fos-lir in the olfactory structures (olfactory bulbs, piriform cortex, and olfactory tubercle), in medial and cortical nuclei of the amygdala, but not in anterior MPOA. Zinc Sulfate 33-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 8730515-7 1996 Stimulation of c-fos by v-Src in growing cells, however, coincided with formation of a complex with an oligonucleotide spanning the c-Sis-inducible element (SIE) upstream from the SRE, suggesting that the signal transduction and activator of transcription (STAT) family of transcription factors, which bind here, may function in response to the v-Src oncoprotein. Oligonucleotides 103-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 8758711-0 1996 Studies of Qingyangshen (II): modulatory effect of co-treatment with qingyangshen and diphenylhydantoin sodium on rat hippocampal c-fos expression during seizures. qingyangshen 69-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 8758711-0 1996 Studies of Qingyangshen (II): modulatory effect of co-treatment with qingyangshen and diphenylhydantoin sodium on rat hippocampal c-fos expression during seizures. Phenytoin 86-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 8758711-2 1996 In this experiment, we examined the modulatory effect of Qingyangshen (QYS), a traditional Chinese medicine with antiepileptic property, and diphenylhydantoin sodium (DPH) on hippocampal c-fos expression during seizures. Phenytoin 141-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 187-192 8758711-2 1996 In this experiment, we examined the modulatory effect of Qingyangshen (QYS), a traditional Chinese medicine with antiepileptic property, and diphenylhydantoin sodium (DPH) on hippocampal c-fos expression during seizures. Phenytoin 167-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 187-192 8783205-6 1996 Fos expression was low-to-undetectable in the brains of animals treated with chronic ECS followed by saline and sham ECS animals that had been treated identically, but with no administration of electrocurrent. Sodium Chloride 101-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 8945723-3 1996 The number of IL-1 beta-induced c-fos-positive cells in the PVN and SON was significantly lower in rats pretreated with indomethacin than in vehicle-treated rats. Indomethacin 120-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 8945723-6 1996 The induction of c-fos immunoreactivity by central administration of IL-1 beta was blocked by indomethacin (500 micrograms/rat), except in the CeA. Indomethacin 94-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 8783321-0 1996 Desensitization follows excitation of bladder primary afferents by intravesical capsaicin, as shown by c-fos activation in the rat spinal cord. Capsaicin 80-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 8783321-2 1996 Two hours after capsaicin instillation in the bladder numerous Fos cells occurred in lamina I at T12-L2 and in lamina I, intermediolateral gray matter (ILG) and dorsal commissure (DCM) at L5-S1. Capsaicin 16-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 8783321-4 1996 At this time, instillation of 1% acetic acid into the bladder of capsaicin-treated rats induced considerably fewer Fos cells than in animals that had been instilled only with the vehicle solution for capsaicin. Acetic Acid 33-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-118 8783321-4 1996 At this time, instillation of 1% acetic acid into the bladder of capsaicin-treated rats induced considerably fewer Fos cells than in animals that had been instilled only with the vehicle solution for capsaicin. Capsaicin 65-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-118 8783322-0 1996 Enhanced effects of co-administered dexamethasone and diclofenac on inflammatory pain processing and associated spinal c-Fos expression in the rat. Dexamethasone 36-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 8783322-0 1996 Enhanced effects of co-administered dexamethasone and diclofenac on inflammatory pain processing and associated spinal c-Fos expression in the rat. Diclofenac 54-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 8783322-1 1996 This study determines the effects of dexamethasone versus co-administered dexamethasone and diclofenac, on carrageenan-evoked spinal c-Fos expression and peripheral oedema in the freely moving rat. Dexamethasone 37-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 8783322-1 1996 This study determines the effects of dexamethasone versus co-administered dexamethasone and diclofenac, on carrageenan-evoked spinal c-Fos expression and peripheral oedema in the freely moving rat. Carrageenan 107-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 8783322-4 1996 The total number of control carrageenan-evoked c-Fos-LI neurones in the lumbar spinal cord was 121 +/- 5 labelled neurones per section, segments L4-L5, which were predominantly located in the superficial and deep laminae (41 +/- 3% and 40 +/- 2% of the total number of c-Fos-LI neurones per section, respectively) of the dorsal horn of the spinal cord. Carrageenan 28-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 8783322-4 1996 The total number of control carrageenan-evoked c-Fos-LI neurones in the lumbar spinal cord was 121 +/- 5 labelled neurones per section, segments L4-L5, which were predominantly located in the superficial and deep laminae (41 +/- 3% and 40 +/- 2% of the total number of c-Fos-LI neurones per section, respectively) of the dorsal horn of the spinal cord. Carrageenan 28-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 269-274 8783322-7 1996 The effects of dexamethasone on the total number of c-Fos-LI neurones and the peripheral oedema were positively correlated. Dexamethasone 15-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 9772672-0 1996 Effects of ginsenoside Rg1 on c-fos gene expression and cAMP levels in rat hippocampus. Ginsenosides 11-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 8730512-5 1996 Potentiation between KCl and 8-Br-c-AMP was also seen with c-fos messenger ribonucleic acid (mRNA) expression. Potassium Chloride 21-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 8730512-5 1996 Potentiation between KCl and 8-Br-c-AMP was also seen with c-fos messenger ribonucleic acid (mRNA) expression. 8-br-c-amp 29-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 8593811-0 1996 Nicotine-induced cFos expression in the hypothalamic paraventricular nucleus is dependent on brainstem effects: correlations with cFos in catecholaminergic and noncatecholaminergic neurons in the nucleus tractus solitarius. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-21 8593811-0 1996 Nicotine-induced cFos expression in the hypothalamic paraventricular nucleus is dependent on brainstem effects: correlations with cFos in catecholaminergic and noncatecholaminergic neurons in the nucleus tractus solitarius. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-134 8593811-3 1996 The results showed that the magnitude of cFos expression was dependent on the dose of nicotine in all regions studied (P < 0.0006); however, at the two lowest doses, only the NTS and CRH-containing region of the PVN expressed cFos, whereas the LC and the rest of the PVN were activated only by higher doses. Nicotine 86-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-45 8593811-4 1996 Nicotine also elicited a dose-dependent increase in cFos expression in the TH+ neurons of the NTS, with C2 more sensitive than A2. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-56 8593811-6 1996 Fourth ventricular mecamylamine completely blocked nicotine-induced cFos expression throughout the NTS, as well as the PVN. Mecamylamine 19-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-72 8593811-6 1996 Fourth ventricular mecamylamine completely blocked nicotine-induced cFos expression throughout the NTS, as well as the PVN. Nicotine 51-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-72 8714707-1 1996 Using an antibody that recognizes the products of all known members of the fos family of immediate early genes, it was demonstrated that destruction of the nigrostriatal pathway by 6-hydroxydopamine (6-OHDA) lesions of the medial forebrain bundle produces a prolonged (>3 months) elevation of Fos-like immunoreactivity in the striatum. Oxidopamine 181-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 8714707-1 1996 Using an antibody that recognizes the products of all known members of the fos family of immediate early genes, it was demonstrated that destruction of the nigrostriatal pathway by 6-hydroxydopamine (6-OHDA) lesions of the medial forebrain bundle produces a prolonged (>3 months) elevation of Fos-like immunoreactivity in the striatum. Oxidopamine 181-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 296-299 8714707-1 1996 Using an antibody that recognizes the products of all known members of the fos family of immediate early genes, it was demonstrated that destruction of the nigrostriatal pathway by 6-hydroxydopamine (6-OHDA) lesions of the medial forebrain bundle produces a prolonged (>3 months) elevation of Fos-like immunoreactivity in the striatum. Oxidopamine 200-206 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 8714707-1 1996 Using an antibody that recognizes the products of all known members of the fos family of immediate early genes, it was demonstrated that destruction of the nigrostriatal pathway by 6-hydroxydopamine (6-OHDA) lesions of the medial forebrain bundle produces a prolonged (>3 months) elevation of Fos-like immunoreactivity in the striatum. Oxidopamine 200-206 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 296-299 8714707-3 1996 In the present study, Western blots were performed on nuclear extracts from the intact and denervated striatum of 6-OHDA-lesioned rats to determine the nature of Fos-immunoreactive protein(s) responsible for this increase. Oxidopamine 114-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-165 8714707-4 1996 Approximately 6 weeks after the 6-OHDA lesion, expression of two Fos-related antigens with apparent molecular masses of 43 and 45 kDa was enhanced in the denervated striatum. Oxidopamine 32-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 8714707-5 1996 Chronic haloperidol administration also selectively elevated expression of these Fos-related antigens, suggesting that their induction after dopaminergic denervation is mediated by reduced activation of D2-like dopamine receptors. Haloperidol 8-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-84 8714707-6 1996 Western blot immunostaining using an antibody which recognizes the N-terminus of FosB indicated that the 43 and 45 kDa Fos-related antigens induced by dopaminergic denervation and chronic haloperidol administration may be related to a truncated form of FosB known as deltaFosB. Haloperidol 188-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-84 8721156-0 1996 Time course of striatal changes induced by 6-hydroxydopamine lesion of the nigrostriatal pathway, as studied by combined evaluation of rotational behaviour and striatal Fos expression. Oxidopamine 43-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-172 8721156-2 1996 Apomorphine at a low dosage (0.25 mg/kg) induced contraversive rotation and supersensitive striatal Fos expression that were detected 24-48 h post-lesion and gradually increased in magnitude. Apomorphine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-103 8721156-3 1996 Twenty-four hours after lesion, both high (5 mg/kg) and low doses (0.5 mg/kg) of D-amphetamine induced contraversive rotation and intense striatal Fos activation on the denervated side; however, only the higher dose induced Fos on the normal side. Dextroamphetamine 81-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-150 8721156-3 1996 Twenty-four hours after lesion, both high (5 mg/kg) and low doses (0.5 mg/kg) of D-amphetamine induced contraversive rotation and intense striatal Fos activation on the denervated side; however, only the higher dose induced Fos on the normal side. Dextroamphetamine 81-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 224-227 8721156-5 1996 In the normal striatum, only high doses of amphetamine induced Fos, but Fos induction in the denervated striatum was similar with both doses: areas showing severely decreased TH immunoreactivity still showed considerable Fos immunoreactivity, and some areas still showing TH immunoreactivity had higher Fos density than in the normal side. Amphetamine 43-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 8721156-6 1996 Seven and 14 days after lesion the loss of TH immunoreactivity and apomorphine-induced supersensitive Fos expression were more evenly distributed, and amphetamine induced only ipsiversive rotation and a low density of Fos-positive nuclei in the denervated striatum. Apomorphine 67-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 8721156-6 1996 Seven and 14 days after lesion the loss of TH immunoreactivity and apomorphine-induced supersensitive Fos expression were more evenly distributed, and amphetamine induced only ipsiversive rotation and a low density of Fos-positive nuclei in the denervated striatum. Amphetamine 151-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 218-221 8591983-11 1996 In addition, ATP stimulated the expression of a 62 kDa c-fos dependent protein in a dose- and time-dependent manner. Adenosine Triphosphate 13-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 8591983-12 1996 Our results thus suggest that extracellular ATP, in the presence of insulin, may be a cofactor in the regulation of thyroid cell proliferation, probably by phosphorylating MAP kinase and stimulating the expression of c-fos. Adenosine Triphosphate 44-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 217-222 8848168-9 1996 Nevertheless, expression of c-Fos is not sufficient for survival since phorbol esters induce c-Fos with no effect on survival. Phorbol Esters 71-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 8821828-0 1996 Inhibition of mitogenesis and c-fos induction in mesangial cells by heparin and heparan sulfates. Heparin 68-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 8821828-0 1996 Inhibition of mitogenesis and c-fos induction in mesangial cells by heparin and heparan sulfates. Heparitin Sulfate 80-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 8821828-3 1996 Added at the time of serum stimulation, heparin (1 microgram/ml or less) causes a decrease in the subsequent expression of c-fos mRNA in RMC, and a similar effect is observed with heparan sulfate chains isolated from RMC-cultures themselves. Heparin 40-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 8821828-6 1996 The effect of heparin on c-fos induction may be independent of interaction with cytokines or cytokine receptors; its magnitude is not diminished when heparin-binding substances are removed from serum by heparin-Sepharose. Heparin 14-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 8821828-6 1996 The effect of heparin on c-fos induction may be independent of interaction with cytokines or cytokine receptors; its magnitude is not diminished when heparin-binding substances are removed from serum by heparin-Sepharose. Heparin 150-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 8821828-6 1996 The effect of heparin on c-fos induction may be independent of interaction with cytokines or cytokine receptors; its magnitude is not diminished when heparin-binding substances are removed from serum by heparin-Sepharose. Heparin 150-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 8821828-6 1996 The effect of heparin on c-fos induction may be independent of interaction with cytokines or cytokine receptors; its magnitude is not diminished when heparin-binding substances are removed from serum by heparin-Sepharose. Sepharose 211-220 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 8821828-7 1996 Furthermore, direct activation of protein kinase C (PKC) with a phorbol ester in the absence of serum likewise induces c-fos and 1 microgram/ml heparin inhibits this response by 65%. Phorbol Esters 64-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 8821828-10 1996 When PKC was inhibited with staurosporine, only very low levels of c-fos were induced by serum. Staurosporine 28-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 8821828-11 1996 We conclude that low concentrations of heparin and heparan sulfate suppress the mitogenic response of mesangial cells to serum and inhibit c-fos mRNA induction through an effect of cell surface-bound glycosaminoglycan on a signalling pathway downstream of PKC. Heparin 39-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 8821828-11 1996 We conclude that low concentrations of heparin and heparan sulfate suppress the mitogenic response of mesangial cells to serum and inhibit c-fos mRNA induction through an effect of cell surface-bound glycosaminoglycan on a signalling pathway downstream of PKC. Heparitin Sulfate 51-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 8821836-7 1996 Phosphorus restriction prevented increases in the expression of ODC, c-fos and c-jun observed in uremia. Phosphorus 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 8821836-9 1996 A single injection of 1,25-(OH)2D3 to uremic rats caused an increase in the steady-state calbindin-D9k mRNA level, and decreases in steady state c-fos and ODC mRNA levels, suggesting that the deficiency of 1,25-(OH)2D3 is responsible for intestinal dysfunction in uremia. Calcitriol 22-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-150 8822533-0 1996 Effects of olanzapine on regional C-Fos expression in rat forebrain. Olanzapine 11-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 8822533-1 1996 Compared to typical antipsychotic drugs, clozapine produces a unique pattern of Fos-like immunoreactive neurons in the rat forebrain. Clozapine 41-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 8822533-3 1996 In the present study, we examined the ability of olanzapine to increase the number of Fos-like immunoreactive neurons in the striatum, nucleus accumbens, lateral septal nucleus, and prefrontal cortex. Olanzapine 49-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 8822533-4 1996 Olanzapine (5, 10 mg/kg) produced dose-dependent increases in the number of Fos-positive neurons in the nucleus accumbens and lateral septal nucleus, important components of the limbic system that may mediate some of the therapeutic actions of neuroleptics. Olanzapine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 8822533-5 1996 Olanzapine also produced dose-dependent increases in the number of Fos-positive neurons in the dorsolateral striatum, an effect that correlates with the ability of neuroleptics to produce extrapyramidal side-effects. Olanzapine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 8822533-6 1996 The effects of olanzapine on regional c-fos expression are not therefore identical to clozapine, which is without effect in the dorsolateral striatum. Olanzapine 15-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 8822533-8 1996 Olanzapine also increased the number of Fos-positive neurons in medical prefrontal cortex, an action unique to clozapine and a few other atypical antipsychotics. Olanzapine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 8822533-8 1996 Olanzapine also increased the number of Fos-positive neurons in medical prefrontal cortex, an action unique to clozapine and a few other atypical antipsychotics. Clozapine 111-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 10627219-0 1996 Suppression of noxious stimulus-evoked expression of Fos protein-like immunoreactivity in rat spinal cord by a selective cannabinoid agonist. Cannabinoids 121-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 10627219-3 1996 Therefore, c-fos immunocytochemistry was used to explore the possibility that cannabinoids reduce behavioral responses to noxious stimuli by decreasing spinal processing of nociceptive inputs. Cannabinoids 78-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 10627219-7 1996 Immunocytochemical processing of sections with an antibody to the Fos protein revealed that the cannabinoid markedly suppressed pain-evoked c-fos expression in the superficial and neck regions of the spinal dorsal horn, but not in the nucleus proprius. Cannabinoids 96-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 10627219-7 1996 Immunocytochemical processing of sections with an antibody to the Fos protein revealed that the cannabinoid markedly suppressed pain-evoked c-fos expression in the superficial and neck regions of the spinal dorsal horn, but not in the nucleus proprius. Cannabinoids 96-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 9045076-3 1996 C-fos messenger RNA levels were also increased throughout hippocampal and cortical subfields following (1S,3R)-1-aminocyclopentane-1,3-dicarboxylic acid administration. 1-amino-1,3-dicarboxycyclopentane 103-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9045076-4 1996 (1S,3R)-1-Aminocyclopentane-1,3-dicarboxylic acid-induced changes in messenger RNA levels occurred without behavioral seizures, yet these changes were similar in magnitude and time course to early changes in neurotrophin and c-fos messenger RNA levels observed following recurrent limbic seizures. 1-amino-1,3-dicarboxycyclopentane 0-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 225-230 9045076-7 1996 These results suggest that (1S,3R)-1-aminocyclopentane-1,3-dicarboxylic acid-sensitive metabotropic glutamate receptors can dramatically increase the expression of neurotrophin and c-fos messenger RNAs in rat forebrain without producing significant behavioral trauma and that these influences may involve ionotropic glutamate receptors in certain brain regions. 1-amino-1,3-dicarboxycyclopentane 27-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 181-186 8758693-2 1996 It was found that a remarkable expression of c-fos was induced by pressure overload and the expression was greatly attenuated by angiotensin converting enzyme inhibitor captopril. Captopril 169-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 8866715-2 1996 The effects of selective adenosine A1 and A2a receptor agonists on locomotion and c-Fos induction following a moderate dose of amphetamine was assessed in rats. Amphetamine 127-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 8919295-2 1996 administration of beta-endorphin and naloxone, an opioid antagonist, on the induction of c-fos and corticotropin-releasing factor (CRF) mRNA to clarify the effects of beta-endorphin on cellular activity and CRF gene expression in the paraventricular nucleus (PVN) of the rat using in situ hybridization. Naloxone 37-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 8551356-8 1996 In addition to reversing the changes in immune response, intrathecal administration of MK-801 reversed the pattern of c-fos immunoreactivity in the spinal cord after immune challenge in neuropathic animals. Dizocilpine Maleate 87-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 8720501-3 1996 To this end, stress-induced c-fos expression was characterized in adrenalectomized (ADX) or adrenalectomized and corticosterone replaced (ADX/B) male rats. Corticosterone 113-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 23888922-2 1996 On gestational day 14, acute administration of terbutaline to pregnant rats resulted in sixfold induction of c-fos mRNA within 1 h; the same increase was obtained when a membrane-permeable analogue of cAMP was given. Terbutaline 47-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 23888922-3 1996 Treatment with dexamethasone on gestational days 11,12 and 13 produced the same increase in c-fos mRNA on gestational day 14 as had been seen with acute terbutaline or CAMP; no further increase could be obtained with acute CAMP treatment in the dexamethasone-pretreated animals. Dexamethasone 15-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 23888922-3 1996 Treatment with dexamethasone on gestational days 11,12 and 13 produced the same increase in c-fos mRNA on gestational day 14 as had been seen with acute terbutaline or CAMP; no further increase could be obtained with acute CAMP treatment in the dexamethasone-pretreated animals. Terbutaline 153-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 8736578-1 1996 A marked expression of the c-fos proto-oncogene has been recently reported in cells of the anterior lobe of the pituitary gland in rats subject to electroacupuncture or noxious thermal stimulation under pentobarbital anaesthesia. Pentobarbital 203-216 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 8866715-5 1996 Pretreatment with the adenosine A2a receptor agonist APEC, but not the adenosine A1 receptor agonist CHA, attenuated c-Fos induction in caudate-putamen and nucleus accumbens by amphetamine. Amphetamine 177-188 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 8866715-6 1996 These findings indicate that amphetamine-induced behavior is subject to modulation by adenosine receptors through mechanisms which are both related to and independent of c-Fos induction. Amphetamine 29-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-175 8866694-1 1996 The expression of proteins coded by the immediate early genes of the fos family and c-jun was used to study the effect of acute ethanol administration on convulsant-induced neuronal activity in rat brain. Ethanol 128-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 8866694-4 1996 Acute IP doses of ethanol (1.0-3.0 g/kg) significantly reduced the behaviours and FOS-immunoreactivity induced in the cerebral cortex by both pentylenetetrazole and N-methyl-D-aspartic acid. Ethanol 18-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 8866694-4 1996 Acute IP doses of ethanol (1.0-3.0 g/kg) significantly reduced the behaviours and FOS-immunoreactivity induced in the cerebral cortex by both pentylenetetrazole and N-methyl-D-aspartic acid. Pentylenetetrazole 142-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 8866694-4 1996 Acute IP doses of ethanol (1.0-3.0 g/kg) significantly reduced the behaviours and FOS-immunoreactivity induced in the cerebral cortex by both pentylenetetrazole and N-methyl-D-aspartic acid. N-Methylaspartate 165-189 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 8866694-5 1996 Pentylenetetrazole-induced FOS-immunoreactivity in the hippocampus was also inhibited by ethanol. Pentylenetetrazole 0-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 8866694-5 1996 Pentylenetetrazole-induced FOS-immunoreactivity in the hippocampus was also inhibited by ethanol. Ethanol 89-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 8866694-6 1996 In contrast, N-methyl-D-aspartic acid-induced FOS-immunoreactivity in the hippocampus was not inhibited by any dose of ethanol. N-Methylaspartate 13-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 8825185-0 1996 Cytochrome P4502E1- and cytochrome P4502B1/2B2-catalyzed carbon tetrachloride metabolism: effects on signal transduction as demonstrated by altered immediate-early (c-Fos and c-Jun) gene expression and nuclear AP-1 and NF-kappa B transcription factor levels. Carbon Tetrachloride 57-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 8717354-0 1996 Caffeine-induced expression of c-fos mRNA and NGFI-A mRNA in caudate putamen and in nucleus accumbens are differentially affected by the N-methyl-D-aspartate receptor antagonist MK-801. Caffeine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 8717354-0 1996 Caffeine-induced expression of c-fos mRNA and NGFI-A mRNA in caudate putamen and in nucleus accumbens are differentially affected by the N-methyl-D-aspartate receptor antagonist MK-801. Dizocilpine Maleate 178-184 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 8717354-6 1996 The two NMDA receptor antagonists significantly reduced the caffeine-induced expression of both c-fos mRNA and NGFI-A mRNA in the medial part of the caudate putamen. Caffeine 60-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 8717354-8 1996 MK-801 caused an enhancement of c-fos and NGFI-A mRNA expression in nucleus accumbens. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 8717354-10 1996 These findings suggest that glutamatergic transmission via NMDA receptors contributes to the induction of c-fos mRNA and NGFI-A mRNA by caffeine in striatum. Caffeine 136-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 8825185-1 1996 We have previously reported that carbon tetrachloride (CCI4) stimulates c-fos, c-jun, and Ca(2+)-activated neutral protease gene expression in rat hepatic tissue (Zawaski et al., Biochem. cci4 55-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 8602107-6 1996 As with the expression of c-fos, supersensitive DI receptors may play a key role in the enhanced induction of AP-I and CREB DNA-binding activity in the dopamine-depleted striatum. Dopamine 152-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 8602107-7 1996 Supershift analysis revealed that c-Fos; and Jun family proteins are the main components for AP-1 induced in the dopamine-depleted striaturn by SKF38393 or levodopa. Dopamine 113-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 8602107-7 1996 Supershift analysis revealed that c-Fos; and Jun family proteins are the main components for AP-1 induced in the dopamine-depleted striaturn by SKF38393 or levodopa. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 144-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 8602107-7 1996 Supershift analysis revealed that c-Fos; and Jun family proteins are the main components for AP-1 induced in the dopamine-depleted striaturn by SKF38393 or levodopa. Levodopa 156-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 8838143-0 1996 Phorbol ester inhibition of rat gonadotropin-releasing hormone promoter activity: role of Fos and Jun in the repression of transcription. Phorbol Esters 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 8838143-3 1996 Treatment of cells with TPA increased c-fos mRNA 20-fold with only a 2-fold increase in c-jun mRNA levels. Tetradecanoylphorbol Acetate 24-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 8786801-4 1996 The levels of c-fos mRNA in the hippocampus and cortex of 3-month-old rats returned to control levels by 3 h after PTZ administration. Pentylenetetrazole 115-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 8786801-5 1996 The levels of c-fos mRNA in the hippocampus and cortex of 20-month-old and 30-month-old rats peaked at 3 h and returned to basal levels by 15 h following PTZ treatment. Pentylenetetrazole 154-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 9116697-0 1996 C-fos expression in the brainstem after voluntary ingestion of sucrose in the rat. Sucrose 63-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 9116697-2 1996 The distribution of the evoked expression of c-fos was visualized in the rat brainstem after voluntary ingestion of sucrose. Sucrose 116-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 9116697-3 1996 Sucrose-taste-specific c-fos immunoreactive neurons were found within the medial part of the nucleus of the solitary tract, the dorsal motor nucleus of the vagus at caudal levels, the paratrigeminal nucleus and the lateral and medial part of the parabrachial nucleus. Sucrose 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 8735971-1 1996 In this study, we carried out an immunohistochemical evaluation of the neurochemical characteristics of neurons that are activated (i.e., express Fos protein) in response to systemic administration of nitroglycerin. Nitroglycerin 201-214 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-149 9259051-3 1996 We have now found in rat aorta that carbachol, a muscarinic cholinergic agonist that promotes release of nitric oxide (NO), inhibits expression of c-fos and c-jun mRNA induced by alpha 1 receptors. Carbachol 36-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-152 9259051-4 1996 NO synthase inhibitors blocked the effects of carbachol on c-fos mRNA and a cGMP analog mimicked carbachol. Carbachol 46-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 9259051-5 1996 After balloon injury in rat aorta using in situ hybridization, the catecholamine-induced increase in c-fos mRNA expression in the medial layer was inhibited by the alpha 1 receptor antagonists, prazosin and chloroethylclonidine. Catecholamines 67-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 9259051-5 1996 After balloon injury in rat aorta using in situ hybridization, the catecholamine-induced increase in c-fos mRNA expression in the medial layer was inhibited by the alpha 1 receptor antagonists, prazosin and chloroethylclonidine. Prazosin 194-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 9259051-5 1996 After balloon injury in rat aorta using in situ hybridization, the catecholamine-induced increase in c-fos mRNA expression in the medial layer was inhibited by the alpha 1 receptor antagonists, prazosin and chloroethylclonidine. chlorethylclonidine 207-227 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 9259051-7 1996 These results suggest that NO, acting through a cGMP-dependent mechanism, inhibits expression of the c-fos and c-jun genes in arteries, which may contribute to the growth-inhibiting effects of the endothelium. Cyclic GMP 48-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 8742472-1 1995 We have used in situ hybridization to investigate the modulation of expression of the immediate early genes (IEGs) c-fos, fos-B, zif/268 and CREM in rat brain following oral administration of saccharin, i.p. Saccharin 192-201 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 8742472-3 1995 Modulation of c-fos, zif/268 and CREM was detected in the NTS, PBN, hypothalamic PVN and central nucleus of the amygdala after the administration of the UCS but not the CS. Cesium 154-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 8750823-0 1995 Spontaneous and evoked glutamate signalling influences Fos-lacZ expression and pyramidal cell death in hippocampal slice cultures from transgenic rats. Glutamic Acid 23-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 8750823-5 1995 An association was observed between cell death, as determined by propidium iodide (PI) staining, and Fos-lacZ expression. Propidium 65-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-104 8750823-7 1995 Long-term treatment with TTX, CNQX, or D,L-APV inhibited the spontaneous neuronal death as well as Fos-lacZ expression. Tetrodotoxin 25-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 8750823-7 1995 Long-term treatment with TTX, CNQX, or D,L-APV inhibited the spontaneous neuronal death as well as Fos-lacZ expression. 6-Cyano-7-nitroquinoxaline-2,3-dione 30-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 8750823-7 1995 Long-term treatment with TTX, CNQX, or D,L-APV inhibited the spontaneous neuronal death as well as Fos-lacZ expression. d,l-apv 39-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 8750823-8 1995 Furthermore, Fos-lacZ induction and cell death could be evoked by removal of these receptor antagonists or by application of the excitotoxin, kainic acid. Kainic Acid 142-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 8750832-1 1995 The dopamine receptor antagonist, haloperidol, produced a time-dependent differential induction of inducible transcription factors (ITFs) in rat striatal neurons: Fos, Fos B, Jun B, Jun D, Krox 20, and Krox 24, but not c-Jun, were induced in the caudate putamen and nucleus accumbens with varying time courses. Haloperidol 34-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 8750832-2 1995 The induction of Fos by haloperidol was stronger in anterior versus posterior regions of the striatum. Haloperidol 24-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 8750832-3 1995 In contrast, induction of Fos by the muscarinic agonist pilocarpine was stronger in the posterior regions of the striatum suggesting that muscarinic receptors do not play a role in the induction of ITFs in striatal neurons by haloperidol. Pilocarpine 56-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 8821738-0 1995 When is the maximal effect of pre-administered systemic morphine on carrageenin evoked spinal c-Fos expression in the rat? Morphine 56-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 8821738-0 1995 When is the maximal effect of pre-administered systemic morphine on carrageenin evoked spinal c-Fos expression in the rat? Carrageenan 68-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 8821738-6 1995 Systemic morphine did not significantly decrease the number of superficial Fos-LI neurons observed 1 h after carrageenin, whereas it significantly reduced the number of superficial Fos-LI neurons induced at 1.5 h and 2 h after carrageenin (58 +/- 3% and 57 +/- 10% reduction, P < 0.001, respectively). Morphine 9-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 181-184 8821738-7 1995 In addition, morphine reduced the number of deep Fos-LI neurons at 1.5 h and 2 h after carrageenin (86 +/- 4%, P < 0.01 and 82 +/- 8%, P < 0.001 reduction as compared to control carrageenin expression, respectively). Morphine 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 8821738-9 1995 Thus, the peak effect of pre-administered morphine on carrageenin evoked c-Fos expression was observed 1.5 h and 2 h after intraplantar carrageenin, with a weaker effect observed at 2.5 h after carrageenin. Morphine 42-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 8821738-9 1995 Thus, the peak effect of pre-administered morphine on carrageenin evoked c-Fos expression was observed 1.5 h and 2 h after intraplantar carrageenin, with a weaker effect observed at 2.5 h after carrageenin. Carrageenan 54-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 8821738-9 1995 Thus, the peak effect of pre-administered morphine on carrageenin evoked c-Fos expression was observed 1.5 h and 2 h after intraplantar carrageenin, with a weaker effect observed at 2.5 h after carrageenin. Carrageenan 136-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 8821738-9 1995 Thus, the peak effect of pre-administered morphine on carrageenin evoked c-Fos expression was observed 1.5 h and 2 h after intraplantar carrageenin, with a weaker effect observed at 2.5 h after carrageenin. Carrageenan 136-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 8750961-6 1995 Acute administration of L-DOPA to DA-denervated animals elicited contralateral rotational behavior as well as a pronounced c-fos protein immunoreactivity in the striatum ipsilateral to the lesion. Levodopa 24-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 8719628-3 1995 In intact rats, the D1 agonist SKF 38393 (20.0 mg/kg) produced a five-fold potentiation of the GP Fos expression due to the D2 agonist quinpirole produced significant Fos expression. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 31-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 8719628-3 1995 In intact rats, the D1 agonist SKF 38393 (20.0 mg/kg) produced a five-fold potentiation of the GP Fos expression due to the D2 agonist quinpirole produced significant Fos expression. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 31-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 167-170 8719628-3 1995 In intact rats, the D1 agonist SKF 38393 (20.0 mg/kg) produced a five-fold potentiation of the GP Fos expression due to the D2 agonist quinpirole produced significant Fos expression. Quinpirole 135-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 8719628-3 1995 In intact rats, the D1 agonist SKF 38393 (20.0 mg/kg) produced a five-fold potentiation of the GP Fos expression due to the D2 agonist quinpirole produced significant Fos expression. Quinpirole 135-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 167-170 8719629-8 1995 The induction of c-fos mRNA and Fos protein in neurons of the STN and SNr following IA-lesions of the CN and GP was reduced markedly by non-NMDA receptor antagonist (GYKI52466), but not by NMDA receptor antagonist (MK-801). Dizocilpine Maleate 215-221 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 8719629-8 1995 The induction of c-fos mRNA and Fos protein in neurons of the STN and SNr following IA-lesions of the CN and GP was reduced markedly by non-NMDA receptor antagonist (GYKI52466), but not by NMDA receptor antagonist (MK-801). Dizocilpine Maleate 215-221 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 8825185-10 1996 Treatment of animals with CCI4 increased c-fos and c-jun mRNA levels from below the limit of detection in control tissue to intense bands within 30 min of treatment, with maximal expression monitored at 1 and 2 hr posttreatment. cci4 26-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 8825185-11 1996 Treatment of animals with diallyl sulfide alone also elevated c-fos and c-jun mRNA expression to detectable levels. allyl sulfide 26-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 8825185-12 1996 However, pretreatment of animals with diallyl sulfide before treatment with CCI4 produced a 76-92% decrease in c-fos and c-jun mRNA levels, relative to that monitored for CCI4-treated animals. allyl sulfide 38-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 8825185-12 1996 However, pretreatment of animals with diallyl sulfide before treatment with CCI4 produced a 76-92% decrease in c-fos and c-jun mRNA levels, relative to that monitored for CCI4-treated animals. cci4 76-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 8825185-13 1996 Pretreatment with N-acetylcysteine did not affect c-fos or c-jun mRNA levels and diminished CCI4-stimulated c-fos and c-jun gene expression by 44 and 55%, respectively, relative to the immediate-early gene mRNA levels monitored in the hepatic tissue of CCI4-treated animals. Acetylcysteine 18-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 8825185-13 1996 Pretreatment with N-acetylcysteine did not affect c-fos or c-jun mRNA levels and diminished CCI4-stimulated c-fos and c-jun gene expression by 44 and 55%, respectively, relative to the immediate-early gene mRNA levels monitored in the hepatic tissue of CCI4-treated animals. cci4 92-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 8825185-14 1996 Pretreatment of animals with the CYP2E1 inducer pyridine for 24 hr had only a marginal effect on c-fos mRNA levels, but increased CCI4-stimulated c-fos and c-jun mRNA levels by an additional approximately 2- to approximately 4-fold over those monitored in the uninduced hepatic tissue of CCI4-treated animals. pyridine 48-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 8825185-14 1996 Pretreatment of animals with the CYP2E1 inducer pyridine for 24 hr had only a marginal effect on c-fos mRNA levels, but increased CCI4-stimulated c-fos and c-jun mRNA levels by an additional approximately 2- to approximately 4-fold over those monitored in the uninduced hepatic tissue of CCI4-treated animals. pyridine 48-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-151 8825185-1 1996 We have previously reported that carbon tetrachloride (CCI4) stimulates c-fos, c-jun, and Ca(2+)-activated neutral protease gene expression in rat hepatic tissue (Zawaski et al., Biochem. Carbon Tetrachloride 33-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 8786852-5 1995 A series of experiments was conducted in order to determine whether this manipulation affected the pattern of Fos immunoreactivity in the basal ganglia elicited by dopamine depletion. Dopamine 164-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 8825185-14 1996 Pretreatment of animals with the CYP2E1 inducer pyridine for 24 hr had only a marginal effect on c-fos mRNA levels, but increased CCI4-stimulated c-fos and c-jun mRNA levels by an additional approximately 2- to approximately 4-fold over those monitored in the uninduced hepatic tissue of CCI4-treated animals. cci4 130-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-151 8825185-15 1996 Whereas phenobarbital treatment alone enhanced c-fos expression only marginally, CCI4 treatment of phenobarbital-pretreated animals increased c-fos expression by up to an additional approximately 8-fold and c-jun mRNA levels by up to an additional approximately 5-fold over the respective levels monitored in the hepatic tissue of CCI4-treated animals. Phenobarbital 8-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 8825185-15 1996 Whereas phenobarbital treatment alone enhanced c-fos expression only marginally, CCI4 treatment of phenobarbital-pretreated animals increased c-fos expression by up to an additional approximately 8-fold and c-jun mRNA levels by up to an additional approximately 5-fold over the respective levels monitored in the hepatic tissue of CCI4-treated animals. cci4 81-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-147 8825185-15 1996 Whereas phenobarbital treatment alone enhanced c-fos expression only marginally, CCI4 treatment of phenobarbital-pretreated animals increased c-fos expression by up to an additional approximately 8-fold and c-jun mRNA levels by up to an additional approximately 5-fold over the respective levels monitored in the hepatic tissue of CCI4-treated animals. Phenobarbital 99-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-147 8825185-16 1996 Enhanced CYP2E1 or CYP2B1/2B2 levels after treatment with pyridine or phenobarbital elevated c-fos mRNA over untreated controls. pyridine 58-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 8825185-16 1996 Enhanced CYP2E1 or CYP2B1/2B2 levels after treatment with pyridine or phenobarbital elevated c-fos mRNA over untreated controls. Phenobarbital 70-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 8846081-7 1995 These results indicated that the transplanted cells continued expressing the c-fos transgene when the rats were given drinking water containing zinc, resulting in the promotion of cell growth and of neovascularization. Water 127-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 8825185-22 1996 Thus, the magnitude of CCI4 stimulation of the immediate-early genes c-fos and c-jun is dependent on metabolic activation by the P450s, and the magnitude of the effect is dependent on the levels and isozyme composition of P450s in the tissue. cci4 23-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 8848114-4 1995 Oxytocin antisense specifically (i) reduced the electrophysiological responses of putative oxytocin, but not vasopressin neurons, (ii) inhibited cholecystokinin-induced and electrically stimulated release of oxytocin from the neurohypophysis, and (iii) reversibly abolished cholecystokinin-induced expression of Fos within the supraoptic nucleus. Cholecystokinin 145-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 312-315 8749800-1 1995 The present study examined fetal alcohol effects (FAE) on the induction of the immediate early genes (IEGs) c-fos, jun B, c-jun, and zif268 mRNAs in the prefrontal cortex, hippocampus, and other brain regions after testing in an alternation task. Alcohols 33-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 7489253-15 1995 Similarly, preincubation of cells with SB for 30 minutes and subsequent incubation in PDGF-BB plus SB diminished PDGF-BB-induced transcription of c-fos, c-jun, and c-myc. Butyric Acid 39-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-151 8925281-0 1995 Attenuation of c-Fos expression in the rat lumbosacral spinal cord by morphine or tramadol following noxious colorectal distention. Morphine 70-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 8925281-0 1995 Attenuation of c-Fos expression in the rat lumbosacral spinal cord by morphine or tramadol following noxious colorectal distention. Tramadol 82-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 8925281-2 1995 This study examined the effects of the analgesics morphine and tramadol on c-Fos expression resulting from noxious CRD in the rat. Morphine 50-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 8925281-2 1995 This study examined the effects of the analgesics morphine and tramadol on c-Fos expression resulting from noxious CRD in the rat. Tramadol 63-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 8925281-4 1995 with morphine (1.25 mg/kg-5.0 mg/kg) or tramadol (1 mg/kg-20 mg/kg) dose-dependently attenuated c-Fos expression to CRD in all areas of the L6-S1 spinal gray matter. Morphine 5-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 8925281-4 1995 with morphine (1.25 mg/kg-5.0 mg/kg) or tramadol (1 mg/kg-20 mg/kg) dose-dependently attenuated c-Fos expression to CRD in all areas of the L6-S1 spinal gray matter. Tramadol 40-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 8925281-8 1995 However, the dose of tramadol that eliminated the visceromotor response (7% of control) reduced the c-Fos expression to 47% of control. Tramadol 21-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 8926038-6 1995 In contrast to the selective induction of c-fos and junB by CGRP and forskolin, ATP led to the accumulation of all four immediate early genes studied, i.e., c-fos, junB, c-jun, and TIS11. Colforsin 69-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 8926038-6 1995 In contrast to the selective induction of c-fos and junB by CGRP and forskolin, ATP led to the accumulation of all four immediate early genes studied, i.e., c-fos, junB, c-jun, and TIS11. Adenosine Triphosphate 80-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-162 8709612-0 1995 Studies of qingyangshen (I): Differential expression of hippocampal c-fos proto-oncogene during kainic acid induced acute and chronic seizures. Kainic Acid 96-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 8596661-7 1995 Fos-immunoreactive nuclei were observed in dorsomedial trigeminal caudalis bilaterally when a restricted area on the tip of the tongue was stimulated with capsaicin, but were located predominantly ipsilaterally following stimulation of the lateral tongue. Capsaicin 155-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 8596661-9 1995 Numbers of Fos-immunoreactive nuclei were significantly increased following nicotine and capsaicin in ventrolateral trigeminal nucleus caudalis and nucleus of the solitary tract. Nicotine 76-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 8596661-9 1995 Numbers of Fos-immunoreactive nuclei were significantly increased following nicotine and capsaicin in ventrolateral trigeminal nucleus caudalis and nucleus of the solitary tract. Capsaicin 89-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 8624709-0 1995 Effects of MK801 on Fos expression in the rat brainstem after unilateral labyrinthectomy. Dizocilpine Maleate 11-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 8624709-8 1995 During the decompensation induced by MK801, Fos-LIR neurons appeared in the contra-MVe, the ipsi-PrH and the bilateral-IOB. Dizocilpine Maleate 37-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 8747141-0 1995 Effects of local anaesthesia on formalin-induced Fos expression in the rat dorsal horn. Formaldehyde 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 8747141-6 1995 Fos-LI was detected in the dorsal horn 2 h after the formalin injection. Formaldehyde 53-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 8741770-0 1995 Intrastriatal DNQX induces rotation and pallidal Fos in the 6-OHDA model of Parkinson"s disease. FG 9041 14-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 8741770-0 1995 Intrastriatal DNQX induces rotation and pallidal Fos in the 6-OHDA model of Parkinson"s disease. Oxidopamine 60-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 8741770-2 1995 When infused into the dopamine-denervated striatum, the AMPA-kainate receptor antagonist DNQX dose-dependently elicited contralateral rotation and ipsilateral Fos immunoreactivity (Fos-IR) in the globus pallidus, a target nucleus of striatal output. Dopamine 22-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-162 8741770-2 1995 When infused into the dopamine-denervated striatum, the AMPA-kainate receptor antagonist DNQX dose-dependently elicited contralateral rotation and ipsilateral Fos immunoreactivity (Fos-IR) in the globus pallidus, a target nucleus of striatal output. FG 9041 89-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-162 8741770-2 1995 When infused into the dopamine-denervated striatum, the AMPA-kainate receptor antagonist DNQX dose-dependently elicited contralateral rotation and ipsilateral Fos immunoreactivity (Fos-IR) in the globus pallidus, a target nucleus of striatal output. FG 9041 89-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 181-184 7478530-4 1995 In Rat6 cells, although both ERK1 and ERK2 are activated in response to mitogens that induce c-fos expression, such as Epidermal Growth Factor (EGF), lysophosphatidic acid (LPA) or serum, expression of v-Raf fails to induce c-fos expression and increase proliferation. lysophosphatidic acid 173-176 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 8581489-3 1995 PEG/water induced c-fos in the central core of SFO and PEG/saline induced c-fos in both the central and peripheral regions. Polyethylene Glycols 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 8719538-0 1995 Aspirin and acetaminophen reduced both Fos expression in rat lumbar spinal cord and inflammatory signs produced by carrageenin inflammation. Aspirin 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 8719538-0 1995 Aspirin and acetaminophen reduced both Fos expression in rat lumbar spinal cord and inflammatory signs produced by carrageenin inflammation. Acetaminophen 12-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 8719538-1 1995 This study, performed in freely moving rats, evaluates the effects of the two most prescribed analgesics, aspirin and acetaminophen, on carrageenin inflammation and the associated c-Fos expression in the rat lumbar spinal cord. Aspirin 106-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 180-185 8719538-1 1995 This study, performed in freely moving rats, evaluates the effects of the two most prescribed analgesics, aspirin and acetaminophen, on carrageenin inflammation and the associated c-Fos expression in the rat lumbar spinal cord. Acetaminophen 118-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 180-185 8719538-2 1995 Maximal dorsal horn c-Fos expression is observed 3 h after carrageenin (6 mg/150 microliters of saline), with Fos-like (Fos-LI) neurones being predominantly located in laminae I-II and V-VI (41 +/- 3% and 39 +/- 5% of the total number of Fos-LI neurones per section for the control group, respectively) of the dorsal horn. Carrageenan 59-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 8719538-2 1995 Maximal dorsal horn c-Fos expression is observed 3 h after carrageenin (6 mg/150 microliters of saline), with Fos-like (Fos-LI) neurones being predominantly located in laminae I-II and V-VI (41 +/- 3% and 39 +/- 5% of the total number of Fos-LI neurones per section for the control group, respectively) of the dorsal horn. Sodium Chloride 96-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 8719538-4 1995 reduced the number of Fos-LI neurones induced by carrageenin-inflammation (28 +/- 2% and 45 +/- 1% reduction, respectively; P < 0.001 for both). Carrageenan 49-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-25 8719538-8 1995 There was a tendency for both aspirin and acetaminophen to have a more pronounced effect on the number of Fos-LI neurones located in deeper laminae, these differential effects being significant for 75 mg/kg of aspirin (P < 0.01) and 150 mg/kg of acetaminophen (P < 0.01). Aspirin 30-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 8719538-8 1995 There was a tendency for both aspirin and acetaminophen to have a more pronounced effect on the number of Fos-LI neurones located in deeper laminae, these differential effects being significant for 75 mg/kg of aspirin (P < 0.01) and 150 mg/kg of acetaminophen (P < 0.01). Acetaminophen 42-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 8719538-8 1995 There was a tendency for both aspirin and acetaminophen to have a more pronounced effect on the number of Fos-LI neurones located in deeper laminae, these differential effects being significant for 75 mg/kg of aspirin (P < 0.01) and 150 mg/kg of acetaminophen (P < 0.01). Aspirin 210-217 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 8719538-10 1995 Furthermore there was a positive correlation between the effects of aspirin and acetaminophen on the number of Fos-LI neurones and the inflammatory signs which developed after carrageenin. Aspirin 68-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 8719538-10 1995 Furthermore there was a positive correlation between the effects of aspirin and acetaminophen on the number of Fos-LI neurones and the inflammatory signs which developed after carrageenin. Acetaminophen 80-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 8719538-10 1995 Furthermore there was a positive correlation between the effects of aspirin and acetaminophen on the number of Fos-LI neurones and the inflammatory signs which developed after carrageenin. Carrageenan 176-187 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 8750890-0 1995 Central glucoprivation evoked by administration of 2-deoxy-D-glucose induces expression of the c-fos gene in a subpopulation of neuropeptide Y neurons in the rat hypothalamus. Deoxyglucose 51-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 8750890-2 1995 To elucidate the hypothalamic mechanism of these phenomena, the induction of c-fos gene expression was examined by in situ hybridization using rats with centrally administered 2DG. Deoxyglucose 176-179 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 7593360-6 1995 Up to 3 h after glucose replenishment, differential temporospatial induction of IEG-coded transcription factors of the FOS, JUN and KROX families were observed in moderately injured neuronal subpopulations, including the majority of dentate granule cells and CA3 neurons. Glucose 16-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-122 7487464-2 1995 METHODS: First, dopamine or vehicle was instilled into a stereotaxically placed intracerebral-ventricular (ICV) cannula with or without D1 (SCH 23390) or D2 (haloperidol) antagonist pretreatment in a rat model, and the effect on hepatic c-fos or c-jun protein expression was investigated. Dopamine 16-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 237-242 7487464-4 1995 RESULTS: Intracerebral-ventricular dopamine treatment increased hepatic c-fos immunoreactive protein but had no effect on hepatic c-jun immunoreactive protein expression. Dopamine 35-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 7487464-5 1995 Pretreatment with SCH 23390 inhibited ICV dopamine treatment-induced hepatic c-fos immunoreactive protein expression. SCH 23390 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 7487464-5 1995 Pretreatment with SCH 23390 inhibited ICV dopamine treatment-induced hepatic c-fos immunoreactive protein expression. Dopamine 42-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 7487464-6 1995 Haloperidol pretreatment synergized with ICV dopamine treatment to overexpress hepatic c-fos protein. Haloperidol 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 7487464-6 1995 Haloperidol pretreatment synergized with ICV dopamine treatment to overexpress hepatic c-fos protein. Dopamine 45-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 7487464-7 1995 Haloperidol treatment significantly increased CLP-induced hepatic c-fos and c-jun protein expression and improved survival following CLP. Haloperidol 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 7487464-9 1995 Treatment with the D2 receptor antagonist, haloperidol, increases sepsis-induced hepatic c-fos and c-jun protein expression and improves survival following peritoneal contamination. Haloperidol 43-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 8581489-3 1995 PEG/water induced c-fos in the central core of SFO and PEG/saline induced c-fos in both the central and peripheral regions. Water 4-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 8581489-3 1995 PEG/water induced c-fos in the central core of SFO and PEG/saline induced c-fos in both the central and peripheral regions. Polyethylene Glycols 55-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 8581489-3 1995 PEG/water induced c-fos in the central core of SFO and PEG/saline induced c-fos in both the central and peripheral regions. Sodium Chloride 59-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 7472514-6 1995 Multiple injections of the NK-1 receptor antagonist, CP-96,345, suppressed stimulus-induced Fos expression in gracile nucleus neurons including thalamic relay neurons. CP96 53-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 7591017-5 1995 Both thromboxane A2 mimetics at a concentration of 10(-6) mol/L induced the expression of c-fos and early growth response gene-1 (egr-1) mRNA, with a maximum at 30 minutes. Thromboxanes 5-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 8552240-7 1995 Fos immunoreactivity was significantly induced in the dorsal striatum following acute and repeated amphetamine. Amphetamine 99-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 8787041-0 1995 Chronic dietary lithium inhibits basal c-fos mRNA expression in rat brain. Lithium 16-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 8787041-9 1995 It was found that basal expression of c-fos was significantly reduced in cortical, hippocampal and hypothalamic brain areas of rats fed dietary lithium for 4 weeks. Lithium 144-151 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 8637618-0 1995 The association of thirst, sodium appetite and vasopressin release with c-fos expression in the forebrain of the rat after intracerebroventricular injection of angiotensin II, angiotensin-(1-7) or carbachol. Carbachol 197-206 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 8637618-1 1995 The effect intracerebroventricular injections of angiotensin II (0.1 nm), angiotensin-(1-7) (1 or 100 nm) and carbachol (500 ng) on c-fos expression was examined in the forebrain of Lister hooded rats. Carbachol 110-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 8593586-0 1995 7-Nitro-indazole, a selective inhibitor of neuronal nitric oxide synthase, reduces formalin evoked c-Fos expression in dorsal horn neurons of the rat spinal cord. 7-nitroindazole 0-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 8593586-0 1995 7-Nitro-indazole, a selective inhibitor of neuronal nitric oxide synthase, reduces formalin evoked c-Fos expression in dorsal horn neurons of the rat spinal cord. Formaldehyde 83-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 8593586-1 1995 The effect of intravenous 7-nitro-indazole (7NI), a selective inhibitor of neuronal nitric oxide synthase, on intraplantar formalin evoked spinal expression of c-Fos was studied. 7-nitroindazole 26-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 8593586-1 1995 The effect of intravenous 7-nitro-indazole (7NI), a selective inhibitor of neuronal nitric oxide synthase, on intraplantar formalin evoked spinal expression of c-Fos was studied. 7-nitroindazole 44-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 8593586-1 1995 The effect of intravenous 7-nitro-indazole (7NI), a selective inhibitor of neuronal nitric oxide synthase, on intraplantar formalin evoked spinal expression of c-Fos was studied. Formaldehyde 123-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 8593586-3 1995 In contrast formalin evoked c-Fos expression in the deep laminae was reduced by 15 mg/kg of 7NI (42 +/- 8% reduction of control, P < 0.05). Formaldehyde 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 8593586-5 1995 Our results implicate a functional relationship between c-Fos expression and nitric oxide, at the spinal level, under inflammatory conditions. Nitric Oxide 77-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 8593588-6 1995 Significant induction of c-fos mRNA was seen with hypoxic exposure as short as 15 min and the effects persisted at 10 h of low pO2 exposure. PO-2 127-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 8593588-7 1995 Hypoxia stimulated transcription from a 356 bp fragment of the c-fos promoter linked to a choloramphenicol acetyl transferase reporter in PC-12 but not in neuroblastoma cells. PC 12 ester 138-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 8574661-0 1995 Nitric oxide induces c-fos gene expression via cyclic AMP response element binding protein (CREB) phosphorylation in rat retinal pigment epithelium. Nitric Oxide 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 8574661-1 1995 We have examined whether nitric oxide (NO) induces the expression of c-fos proto-oncogene and the phosphorylation of cyclic AMP response element binding protein (CREB) in the rat retina. Nitric Oxide 25-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 7559590-3 1995 The induction of c-fos by the calcium ionophore A23187 was reduced by treatment with SNAP or 8-bromo-cGMP. Calcium 30-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 7559590-3 1995 The induction of c-fos by the calcium ionophore A23187 was reduced by treatment with SNAP or 8-bromo-cGMP. Calcimycin 48-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 7559590-3 1995 The induction of c-fos by the calcium ionophore A23187 was reduced by treatment with SNAP or 8-bromo-cGMP. 8-bromocyclic GMP 93-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 8566141-0 1995 Amantadine induces c-fos in rat striatum: reversal with dopamine D1 and NMDA receptor antagonists. Amantadine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 8566141-1 1995 Amantadine (1-aminoadamantane) induced Fos expression in the central, dorsal-medial and ventral-medial part of the striatum. Amantadine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 8566141-1 1995 Amantadine (1-aminoadamantane) induced Fos expression in the central, dorsal-medial and ventral-medial part of the striatum. Amantadine 12-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 8566141-2 1995 The distribution pattern of Fos induced by amantadine was more similar to those seen with dopaminomimetics than with N-methyl-D-aspartate (NMDA) receptor antagonists. Amantadine 43-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 8566141-3 1995 Pretreatment with the dopamine D1 receptor antagonist, SCH23390, and the NMDA receptor antagonist, MK-801, blocked amantadine induction of Fos in the striatum. SCH 23390 55-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 8566141-3 1995 Pretreatment with the dopamine D1 receptor antagonist, SCH23390, and the NMDA receptor antagonist, MK-801, blocked amantadine induction of Fos in the striatum. Dizocilpine Maleate 99-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 8566141-3 1995 Pretreatment with the dopamine D1 receptor antagonist, SCH23390, and the NMDA receptor antagonist, MK-801, blocked amantadine induction of Fos in the striatum. Amantadine 115-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 8566141-4 1995 However, amantadine induction of Fos in the striatum was unaffected by the dopamine D2 receptor antagonist, sulpiride. Amantadine 9-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 8566141-5 1995 These results suggest that amantadine induction of Fos in the rat striatum is related to dopamine D1 and NMDA receptors. Amantadine 27-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 8568687-4 1995 Refeeding of fasted rats induced a transient increase in c-fos mRNA abundance in gastric corpus and antrum that was sixfold within 15 min and declined within 4 h. The response was not mediated by gastrinergic or muscarinic cholinergic mechanisms; it was reduced but not abolished by hexamethonium. Hexamethonium 283-296 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 7487997-0 1995 Nuclear matrix condensation and c-myc and c-fos expression are specifically altered in culture rat hepatocytes after exposure to cyproterone acetate and phenobarbital. Cyproterone Acetate 129-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 7487997-0 1995 Nuclear matrix condensation and c-myc and c-fos expression are specifically altered in culture rat hepatocytes after exposure to cyproterone acetate and phenobarbital. Phenobarbital 153-166 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 7487997-4 1995 C-fos expression and decrease in nuclear fluorescence could be induced by both chemicals, phenobarbital being the lesser potent, whereas c-myc expression was only inducible by cyproterone acetate. Phenobarbital 90-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8552240-8 1995 Fos immunoreactivity in the core of the nucleus accumbens was significantly increased following repeated amphetamine only. Amphetamine 105-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 8552248-7 1995 In many cases both inside and outside the suprachiasmatic nucleus, the Fos-immunoreactive cells appeared to make direct contact with NADPH-diaphorase-stained cells or fibres, but no co-localization of Fos immunoreactivity and NADPH-diaphorase histochemical activity within individual cells was detected. NADP 133-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 8552248-8 1995 Extensive co-distribution of NADPH-diaphorase-stained cells and fibres and cells that express Fos in response to photic stimulation in the suprachiasmatic nucleus region is in line with the hypothesis that nitric oxide participates in the mechanism mediating circadian light signalling in the suprachiasmatic nucleus. Nitric Oxide 206-218 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 7568184-2 1995 While a large body of literature describes the induction of immediate-early genes, including c-fos, fosB, c-jun, junB, zif/268, and krox genes by glutamate and agonists in neurons, very little is known about preexisting transcription factors controlling the induction of such genes. Glutamic Acid 146-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 8732045-3 1995 RESULTS: The c-fos expression evoked by intrathecal injection (it) SP 10 micrograms and sc 5% formaldehyde (For) 150 microL in the hindpaw was densely distributed in the laminae I, II, V, and VI of spinal dorsal horn. Formaldehyde 94-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 8732045-6 1995 SP and For induced c-fos expressions in the spinal cord and the pain responses were reduced by 5-HT and increased by 5-HT depletor fenclonine 300 mg.kg-1. Fenclonine 131-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 8974658-0 1995 Amphetamine induction of c-fos in the nucleus accumbens is not inhibited by glutamate antagonists. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 8974658-1 1995 Systemic administration of relatively high doses of amphetamine or cocaine induces expression of c-fos in the rat striatum and nucleus accumbens. Amphetamine 52-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 8974658-1 1995 Systemic administration of relatively high doses of amphetamine or cocaine induces expression of c-fos in the rat striatum and nucleus accumbens. Cocaine 67-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 8974658-3 1995 Therefore, it was determined if low doses of amphetamine capable of eliciting reward and sensitization increase levels of c-Fos protein in the nucleus accumbens. Amphetamine 45-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 8974658-4 1995 Amphetamine, 1 mg/kg, stimulated locomotor activity and increased the number of nucleus accumbens cells immunohistochemically positive for c-Fos protein to approximately 800 cells per section from a control of approximately 100 cells per section. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 8974658-5 1995 Since glutamate antagonists modify various responses to amphetamine, it was then determined whether activation of glutamate receptors is involved in the induction of c-Fos protein by low doses of amphetamine. Amphetamine 196-207 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-171 8974658-8 1995 The AMPA receptor antagonist NBQX by itself had no effect on locomotor activity but increased slightly the number of cells positive for c-Fos protein in the nucleus accumbens. 2,3-dioxo-6-nitro-7-sulfamoylbenzo(f)quinoxaline 29-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 8974658-9 1995 When given before amphetamine, locomotor activity was completely attenuated, and the extent of c-fos induction was greater than from amphetamine alone. Amphetamine 133-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 8974658-10 1995 We conclude that low doses of amphetamine do increase abundance of c-Fos protein in the nucleus accumbens. Amphetamine 30-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 7561843-3 1995 As both c-fos and SS genes have a cyclic AMP response element (CRE) in their promoters, CRE-binding protein (CREB) phosphorylation in retinal cells was examined with a phospho-CREB-specific antibody. Cyclic AMP 34-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-13 8774955-4 1995 Treatment with the D1-dopamine receptor agonist SKF 38393 (10 mg/kg) increased SCN c-fos gene expression during both day and night on postnatal day (PD) 0, 1, and at night on PD 2, but the c-fos response disappeared by PD 4. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 48-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 8774955-4 1995 Treatment with the D1-dopamine receptor agonist SKF 38393 (10 mg/kg) increased SCN c-fos gene expression during both day and night on postnatal day (PD) 0, 1, and at night on PD 2, but the c-fos response disappeared by PD 4. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 48-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 189-194 7472437-3 1995 Exposure to a saccharin solution (CS) which had previously been paired with lithium chloride (LiCl; US) induced significant c-Fos-like immunoreactivity (c-FLI) in the intermediate zone of the nucleus of the solitary tract (NTS), a response that was quite similar to that displayed following administration of LiCl alone. saccharin solution 14-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 8563722-0 1995 Bacterial lipopolysaccharide-induced changes in FOS protein expression in the rat brain: correlation with thermoregulatory changes and plasma corticosterone. Corticosterone 142-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 7472437-3 1995 Exposure to a saccharin solution (CS) which had previously been paired with lithium chloride (LiCl; US) induced significant c-Fos-like immunoreactivity (c-FLI) in the intermediate zone of the nucleus of the solitary tract (NTS), a response that was quite similar to that displayed following administration of LiCl alone. Cesium 34-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 7472437-3 1995 Exposure to a saccharin solution (CS) which had previously been paired with lithium chloride (LiCl; US) induced significant c-Fos-like immunoreactivity (c-FLI) in the intermediate zone of the nucleus of the solitary tract (NTS), a response that was quite similar to that displayed following administration of LiCl alone. Lithium Chloride 76-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 7472437-3 1995 Exposure to a saccharin solution (CS) which had previously been paired with lithium chloride (LiCl; US) induced significant c-Fos-like immunoreactivity (c-FLI) in the intermediate zone of the nucleus of the solitary tract (NTS), a response that was quite similar to that displayed following administration of LiCl alone. Lithium Chloride 94-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 8568921-12 1995 Effects of staurosporine, 6-TG, and sphingosine on c-fos gene induction with or without NGF were not correlated with the generation of neurites. Staurosporine 11-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 8563722-1 1995 In the present study the regions of the brain showing an increase in the number of FOS protein stained cells 180 min following intravenous saline or bacterial lipopolysaccharide (LPS) treatment were investigated and correlated with changes in body temperature and plasma corticosterone levels. Sodium Chloride 139-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 8568921-12 1995 Effects of staurosporine, 6-TG, and sphingosine on c-fos gene induction with or without NGF were not correlated with the generation of neurites. Thioguanine 26-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 8568921-12 1995 Effects of staurosporine, 6-TG, and sphingosine on c-fos gene induction with or without NGF were not correlated with the generation of neurites. Sphingosine 36-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 8563722-1 1995 In the present study the regions of the brain showing an increase in the number of FOS protein stained cells 180 min following intravenous saline or bacterial lipopolysaccharide (LPS) treatment were investigated and correlated with changes in body temperature and plasma corticosterone levels. bacterial lipopolysaccharide 149-177 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 8563722-1 1995 In the present study the regions of the brain showing an increase in the number of FOS protein stained cells 180 min following intravenous saline or bacterial lipopolysaccharide (LPS) treatment were investigated and correlated with changes in body temperature and plasma corticosterone levels. Corticosterone 271-285 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 8563722-9 1995 Plasma corticosterone was unaffected by the lowest dose of LPS (0.35 micrograms), however the higher doses employed (3.5 and 50 micrograms) caused significant increases in plasma corticosterone which correlated with the increases in the number of FOS stained cells in the pPVN. Corticosterone 179-193 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 247-250 8573242-2 1995 In the present study, we followed the post-LPS time-course of immunocytochemical expression of Fos-like activity in CRH41 neurons whose immunolabeling was enhanced by icv colchicine pretreatment 48 h before the LPS, and CRH41 release in the push-pull cannulated median eminence of free-moving rats, in parallel with the ACTH response. Colchicine 171-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 8573242-2 1995 In the present study, we followed the post-LPS time-course of immunocytochemical expression of Fos-like activity in CRH41 neurons whose immunolabeling was enhanced by icv colchicine pretreatment 48 h before the LPS, and CRH41 release in the push-pull cannulated median eminence of free-moving rats, in parallel with the ACTH response. crh41 116-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 8573242-4 1995 Colchicine strongly inhibited the LPS-induced activation of Fos expression in single-labeled paraventricular neurons. Colchicine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 8544984-1 1995 The immunocytochemical localization of the immediate-early gene c-fos has been used to map sites of neuronal activity in the rat brain associated with 5-hydroxytryptamine function. Serotonin 151-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 8544984-0 1995 Induction of c-fos in rat forebrain by pharmacological manipulation of 5-hydroxytryptamine levels. Serotonin 71-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 8577370-13 1995 The intrathalamic grafts of rats which had received a low dose of apomorphine (0.25 mg/kg) 2 h before perfusion showed clusters of intensely Fos-immunoreactive nuclei throughout the transplant, indicating that these cells had developed dopamine receptors and supersensitivity to dopamine agonists. Apomorphine 66-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 8577490-1 1995 Formalin injection into the hindpaw of rats produces many neurons with c-fos protein-like immunoreactivity (fos-neurons) in the medial 3/4 of the ipsilateral dorsal horn laminae I and II at the junction of 4th and 5th lumbar segments (the sciatic territory). Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 8592640-1 1995 Chemical irritation of the urinary bladder with formalin in the rat induced c-fos protein-like immunoreactivity in more than 80% of substance P receptor-like immunoreactive (SPR-LI) neurons of the dorsal commissural nucleus, sacral parasympathetic nucleus and lamina I in the 6th lumbar and 1st sacral cord segments. Formaldehyde 48-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 7588705-5 1995 Furthermore, in the presence of a phorbol ester, only the clones expressing p37SH2/3 showed increased tyrosine phosphorylation of p62 and c-fos expression. Phorbol Esters 34-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 8535846-3 1995 We examined induction of Fos protein in SCN and IGL regions, in response to cage change, intraperitoneal saline injection, and restraint stress. Sodium Chloride 105-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 8748977-0 1995 Norepinephrine induces expression of c-fos mRNA through the alpha-adrenoceptor in rat aortic rings. Norepinephrine 0-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 8748977-1 1995 We examined whether norepinephrine at pharmacologically relevant doses induces increased expression of c-fos mRNA in rat aortic rings. Norepinephrine 20-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 8748977-2 1995 c-fos mRNA was expressed at norepinephrine concentrations known to cause minimum and maximum contraction of rat aorta in vitro. Norepinephrine 28-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8748977-3 1995 At the concentration known to cause maximum contraction, norepinephrine produced a marked and sustained increase of c-fos mRNA expression. Norepinephrine 57-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 8748977-4 1995 Induction of c-fos was blocked completely by the alpha 1-adrenergic antagonist prazosin, partially by the alpha 2-adrenergic antagonist yohimbine, and not at all by the beta-adrenergic antagonist propranolol. Prazosin 79-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 8748977-4 1995 Induction of c-fos was blocked completely by the alpha 1-adrenergic antagonist prazosin, partially by the alpha 2-adrenergic antagonist yohimbine, and not at all by the beta-adrenergic antagonist propranolol. Yohimbine 136-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 8748977-5 1995 A prazosin inhibition curve showed that 1 nmol/L prazosin inhibited 10 micromol/L norepinephrine induced c-fos expression by 40%. Prazosin 2-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 8748977-5 1995 A prazosin inhibition curve showed that 1 nmol/L prazosin inhibited 10 micromol/L norepinephrine induced c-fos expression by 40%. Prazosin 49-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 8748977-5 1995 A prazosin inhibition curve showed that 1 nmol/L prazosin inhibited 10 micromol/L norepinephrine induced c-fos expression by 40%. Norepinephrine 82-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 8748977-6 1995 At the pharmacologic dose known to cause maximum contraction, norepinephrine induces c-fos mRNA expression through the alpha-adrenoceptor in rat aortic rings. Norepinephrine 62-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 8866763-12 1995 Our immediate early gene (IEG) time-course data with c-fos and zif268 in rats following PCP suggest that a single dose of this antiglutamatergic compound can have an effect in some brain areas which lasts beyond 48 h, an effect which is distinct by IEG and by region. Phencyclidine 88-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 7657802-4 1995 During cardiocyte hypertrophy evoked by endothelin-1, Phenylephrine, or PMA, the steady state level of BNP mRNA increased as rapidly as the "immediate-early" induction of the c-fos gene expression, and reached a maximal level within 1 h. Actinomycin D, a transcriptional inhibitor, completely diminished the response, while the translational blocked with cycloheximide did not inhibit it. Tetradecanoylphorbol Acetate 72-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 175-180 8690853-2 1995 Exposure of unanesthetized rats to oxygen deprivation induces activation of the c-fos gene within the nucleus tractus solitarius, resulting in expression of fos-like immunoreactive protein (Fos). Oxygen 35-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 8690853-2 1995 Exposure of unanesthetized rats to oxygen deprivation induces activation of the c-fos gene within the nucleus tractus solitarius, resulting in expression of fos-like immunoreactive protein (Fos). Oxygen 35-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 8690853-2 1995 Exposure of unanesthetized rats to oxygen deprivation induces activation of the c-fos gene within the nucleus tractus solitarius, resulting in expression of fos-like immunoreactive protein (Fos). Oxygen 35-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 190-193 8690853-3 1995 Prior administration of MK-801, a nonselective antagonist of the N-methyl-D-aspartate (NMDA)-sensitive glutamate receptor (1 or 2 mg/kg), significantly attenuated but did not completely block hypoxia-induced Fos expression. Dizocilpine Maleate 24-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 208-211 8690857-5 1995 In control rats treated with isotonic saline, few Fos-positive neurons were observed. Sodium Chloride 38-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 8690857-6 1995 In contrast, phenylephrine and hydralazine treatments resulted in different, yet reproducible, patterns of Fos expression in the brain, with hydralazine evoking Fos expression in more brain regions than phenylephrine. Phenylephrine 13-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 8690857-6 1995 In contrast, phenylephrine and hydralazine treatments resulted in different, yet reproducible, patterns of Fos expression in the brain, with hydralazine evoking Fos expression in more brain regions than phenylephrine. Phenylephrine 13-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-164 8690857-6 1995 In contrast, phenylephrine and hydralazine treatments resulted in different, yet reproducible, patterns of Fos expression in the brain, with hydralazine evoking Fos expression in more brain regions than phenylephrine. Hydralazine 31-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 8690857-6 1995 In contrast, phenylephrine and hydralazine treatments resulted in different, yet reproducible, patterns of Fos expression in the brain, with hydralazine evoking Fos expression in more brain regions than phenylephrine. Hydralazine 31-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-164 8690857-6 1995 In contrast, phenylephrine and hydralazine treatments resulted in different, yet reproducible, patterns of Fos expression in the brain, with hydralazine evoking Fos expression in more brain regions than phenylephrine. Hydralazine 141-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-164 8690857-7 1995 Brain regions containing Fos-positive neurons in rats treated with hydralazine included nucleus tractus solitarius, area postrema, caudal ventrolateral medulla, rostral ventrolateral medulla, bed nucleus of the stria terminalis, amygdala, paraventricular nucleus, supraoptic nucleus, subfornical organ and the Islands of Calleja. Hydralazine 67-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 8690857-8 1995 The nucleus tractus solitarius, paraventricular nucleus and the amygdala also contained Fos-positive neurons in phenylephrine-treated rats, although the number of Fos-positive neurons was always less than that noted in the hydralazine-treated rats and the location of Fos-positive neurons within these regions tended to differ between treatments. Phenylephrine 112-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 8690857-8 1995 The nucleus tractus solitarius, paraventricular nucleus and the amygdala also contained Fos-positive neurons in phenylephrine-treated rats, although the number of Fos-positive neurons was always less than that noted in the hydralazine-treated rats and the location of Fos-positive neurons within these regions tended to differ between treatments. Phenylephrine 112-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 8690857-8 1995 The nucleus tractus solitarius, paraventricular nucleus and the amygdala also contained Fos-positive neurons in phenylephrine-treated rats, although the number of Fos-positive neurons was always less than that noted in the hydralazine-treated rats and the location of Fos-positive neurons within these regions tended to differ between treatments. Phenylephrine 112-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 8590069-2 1995 The purpose of this study was to examine modulation of neuroleptic induction of NT/N and the proto-oncogene c-fos expression by the GABAA agonist muscimol. gabaa 132-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-113 8590069-2 1995 The purpose of this study was to examine modulation of neuroleptic induction of NT/N and the proto-oncogene c-fos expression by the GABAA agonist muscimol. Muscimol 146-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-113 8590069-5 1995 Expression of NT/N and c-fos mRNA was examined by in situ hybridization using 35S-antisense probes. Sulfur-35 78-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 8590069-7 1995 However, co-administration of muscimol with haloperidol reduced haloperidol-induced increases in NT/N as well as c-fos mRNA in the dorsolateral striatum. Muscimol 30-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-118 8590069-7 1995 However, co-administration of muscimol with haloperidol reduced haloperidol-induced increases in NT/N as well as c-fos mRNA in the dorsolateral striatum. Haloperidol 44-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-118 8590069-7 1995 However, co-administration of muscimol with haloperidol reduced haloperidol-induced increases in NT/N as well as c-fos mRNA in the dorsolateral striatum. Haloperidol 64-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-118 7585834-10 1995 Phorbol 12-myristate 13-acetate also decreased [Ca2+]i transients, caused c-fos induction, and provoked hypertrophy in myocytes. Tetradecanoylphorbol Acetate 0-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 7585834-14 1995 Phorbol 12-myristate 13-acetate did not affect [Ca2+]i or cellular growth in nonmyocytes but did cause c-fos induction. Tetradecanoylphorbol Acetate 0-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 7650059-7 1995 The expression of c-jun, jun B, and c-fos mRNA was inhibited by the endothelin type A receptor (ET)-selective antagonist, BQ-123. cyclo(Trp-Asp-Pro-Val-Leu) 122-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 8544984-3 1995 We now report that such treatment induces a specific anatomical pattern of expression of c-fos in rat forebrain in many limbic, striatal and cortical areas which corresponds well with the distribution of 5-hydroxytryptamine-immunoreactive terminals. Serotonin 204-223 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 8544984-5 1995 Two-day pretreatment with p-chlorophenylalanine (300 mg/kg) prior to tranylcypromine and L-tryptophan resulted in a significant attenuation of Fos-like immunoreactivity in specific brain areas, including the piriform and frontal cortices, nucleus accumbens, caudate-putamen, paraventricular hypothalamus and paraventricular thalamic nucleus. Fenclonine 26-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-146 8544984-5 1995 Two-day pretreatment with p-chlorophenylalanine (300 mg/kg) prior to tranylcypromine and L-tryptophan resulted in a significant attenuation of Fos-like immunoreactivity in specific brain areas, including the piriform and frontal cortices, nucleus accumbens, caudate-putamen, paraventricular hypothalamus and paraventricular thalamic nucleus. Tranylcypromine 69-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-146 8544984-5 1995 Two-day pretreatment with p-chlorophenylalanine (300 mg/kg) prior to tranylcypromine and L-tryptophan resulted in a significant attenuation of Fos-like immunoreactivity in specific brain areas, including the piriform and frontal cortices, nucleus accumbens, caudate-putamen, paraventricular hypothalamus and paraventricular thalamic nucleus. Tryptophan 89-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-146 8544984-7 1995 The results of this study suggest that the elevation in Fos-like immunoreactivity following treatment with tryptophan and a monoamine oxidase inhibitor is mainly due to increased 5-hydroxytryptamine synthesis and release. Tryptophan 107-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 8544984-7 1995 The results of this study suggest that the elevation in Fos-like immunoreactivity following treatment with tryptophan and a monoamine oxidase inhibitor is mainly due to increased 5-hydroxytryptamine synthesis and release. Serotonin 179-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 8544995-0 1995 Effect of administration of high dose intrathecal clonidine or morphine prior to sciatic nerve section on c-Fos expression in rat lumbar spinal cord. Clonidine 50-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 8544995-0 1995 Effect of administration of high dose intrathecal clonidine or morphine prior to sciatic nerve section on c-Fos expression in rat lumbar spinal cord. Morphine 63-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 8544995-1 1995 The effects of moderate and high intrathecal doses of clonidine, an alpha 2 adrenoceptor agonist, or a high dose of morphine on sciatic nerve section-induced expression of c-Fos-like immunoreactivity was studied in laminae I and II of the dorsal horn and laminae VIII and IX of the ventral horn of rat lumbar spinal cord. Clonidine 54-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-177 8544995-1 1995 The effects of moderate and high intrathecal doses of clonidine, an alpha 2 adrenoceptor agonist, or a high dose of morphine on sciatic nerve section-induced expression of c-Fos-like immunoreactivity was studied in laminae I and II of the dorsal horn and laminae VIII and IX of the ventral horn of rat lumbar spinal cord. Morphine 116-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-177 8544995-2 1995 c-Fos-like immunoreactivity was examined by immunohistochemistry in normal rats (group 1), rats implanted with an intrathecal catheter with its tip on the lumbar spinal cord (group 2), injected with 10 micrograms (group 3) or 50 micrograms (group 4) clonidine intrathecally 3 h before being killed. Clonidine 250-259 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8544995-6 1995 The level of c-Fos-like immunoreactivity after 10 or 50 micrograms intrathecal clonidine was similar as in the intrathecal catheter group. Clonidine 79-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 8544995-7 1995 Sciatic nerve section caused a significant ipsilateral increase in c-Fos-like immunoreactivity in the dorsal horn compared to the intact side in rats injected with saline. Sodium Chloride 164-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 8544995-8 1995 Pretreatment with 10 or 10 micrograms clonidine did not reduce sciatic nerve section-induced expression of c-Fos-like immunoreactivity, but instead caused a significant bilateral increase in c-Fos-like immunoreactivity. Clonidine 38-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 191-196 7501282-0 1995 Differential regulation of c-fos, proenkephalin and tyrosine hydroxylase gene expression by metrazole in the hamster adrenal and hippocampus. Pentylenetetrazole 92-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 8532189-4 1995 Increased levels of c-fos and c-jun mRNA were observed in the spinal cord at 40 min of morphine withdrawal. Morphine 87-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 8527740-0 1995 Fenfluramine-induced c-fos in the striatum and hypothalamus: a tract-tracing study. Fenfluramine 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 7589222-0 1995 Chronic fluoxetine or desmethylimipramine treatment alters 5-HT2 receptor mediated c-fos gene expression. Fluoxetine 8-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 7589222-0 1995 Chronic fluoxetine or desmethylimipramine treatment alters 5-HT2 receptor mediated c-fos gene expression. Desipramine 22-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 7477937-3 1995 Administration of kainate to rats caused a profound increase in messenger RNAs of the transcription factor genes c-fos and c-jun in the dentate gyrus, followed by an increase in the level of the transcriptional complex activator protein-1 in hippocampal neurons. Kainic Acid 18-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 7477938-0 1995 A single injection of amphetamine or methamphetamine induces dynamic alterations in c-fos, zif/268 and preprodynorphin messenger RNA expression in rat forebrain. Amphetamine 22-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 7477938-0 1995 A single injection of amphetamine or methamphetamine induces dynamic alterations in c-fos, zif/268 and preprodynorphin messenger RNA expression in rat forebrain. Methamphetamine 37-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 7477938-5 1995 Similarly, methamphetamine induced a different pattern of c-fos and zif/268 messenger RNA induction in sensory/motor cortex, dorsal striatum (caudatoputamen) and ventral striatum (nucleus accumbens) than did amphetamine. Methamphetamine 11-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 7477938-5 1995 Similarly, methamphetamine induced a different pattern of c-fos and zif/268 messenger RNA induction in sensory/motor cortex, dorsal striatum (caudatoputamen) and ventral striatum (nucleus accumbens) than did amphetamine. Amphetamine 15-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 7477939-4 1995 Dopamine (10 nM-5 microM) caused a marked increase in the levels of c-fos mRNA and zif/268 mRNA in cultured striatal neurons, an effect that was reproduced by the D1-like dopamine receptor agonist SKF38393 (10 nM-5 microM). Dopamine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 7477939-4 1995 Dopamine (10 nM-5 microM) caused a marked increase in the levels of c-fos mRNA and zif/268 mRNA in cultured striatal neurons, an effect that was reproduced by the D1-like dopamine receptor agonist SKF38393 (10 nM-5 microM). 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 197-205 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 7477939-6 1995 The D2-like agonist quinpirole did not increase zif/268 mRNA above basal levels at concentrations up to 5 microM, but caused a slight increase in the levels of c-fos mRNA. Quinpirole 20-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 7477939-8 1995 These results suggest that SKF38393 acts on striatal neurons to stimulate c-fos expression predominantly through protein kinase C, but also partially through protein kinase A. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 27-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 7496806-3 1995 Pretreatment with the NMDA receptor blocker MK-801 eliminated closed head injury induced Fos expression in the pyriform cortex and attenuated that seen in the hippocampus. Dizocilpine Maleate 44-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 7496806-4 1995 Similar amounts of Fos expression were observed in urethane anaesthetized lactating and nonlactating rats following closed head injury. Urethane 51-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 8584207-0 1995 Endogenous acetylcholine-induced Fos expression in magnocellular neurosecretory neurons in the supraoptic nucleus of the rat hypothalamus. Acetylcholine 11-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 8584207-1 1995 Induction of c-Fos expression in the supraoptic nucleus (SON) of the rat hypothalamus by endogenous acetylcholine was examined by microinfusion of neostigmine, a cholinesterase inhibitor, into the nucleus to locally accumulate the spontaneously released acetylcholine from the cholinergic terminals in the SON. Acetylcholine 100-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 8584207-1 1995 Induction of c-Fos expression in the supraoptic nucleus (SON) of the rat hypothalamus by endogenous acetylcholine was examined by microinfusion of neostigmine, a cholinesterase inhibitor, into the nucleus to locally accumulate the spontaneously released acetylcholine from the cholinergic terminals in the SON. Neostigmine 147-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 8584207-1 1995 Induction of c-Fos expression in the supraoptic nucleus (SON) of the rat hypothalamus by endogenous acetylcholine was examined by microinfusion of neostigmine, a cholinesterase inhibitor, into the nucleus to locally accumulate the spontaneously released acetylcholine from the cholinergic terminals in the SON. Acetylcholine 254-267 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 8584207-3 1995 Fos-like immunoreactivity was manifested in both the vasopressin neurons and oxytocin neurons following the microinfusion of neostigmine. Neostigmine 125-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 7501282-1 1995 Metrazole (MTZ) induces sequential activation of c-fos, proenkephalin (Penk) and tyrosine hydroxylase (TH) gene expression in the rat adrenal and c-fos and Penk gene expression in the rat hippocampus. Pentylenetetrazole 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 8584207-4 1995 Microinfusion of nicotinic agonist, nicotine, to the SON also induced Fos expression, but mainly in the vasopressin neurons. Nicotine 36-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 7501282-1 1995 Metrazole (MTZ) induces sequential activation of c-fos, proenkephalin (Penk) and tyrosine hydroxylase (TH) gene expression in the rat adrenal and c-fos and Penk gene expression in the rat hippocampus. Pentylenetetrazole 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-151 8584207-5 1995 Microinfusion of muscarinic agonist, carbachol, induced Fos expression as well, but mostly in the oxytocin neurons. Carbachol 37-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 7501282-1 1995 Metrazole (MTZ) induces sequential activation of c-fos, proenkephalin (Penk) and tyrosine hydroxylase (TH) gene expression in the rat adrenal and c-fos and Penk gene expression in the rat hippocampus. Pentylenetetrazole 11-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 7501282-1 1995 Metrazole (MTZ) induces sequential activation of c-fos, proenkephalin (Penk) and tyrosine hydroxylase (TH) gene expression in the rat adrenal and c-fos and Penk gene expression in the rat hippocampus. Pentylenetetrazole 11-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-151 7501282-2 1995 As in the rat, MTZ produced a dose-dependent induction of c-fos mRNA in the hamster adrenal and hippocampus together with an increase in adrenal TH mRNA. Pentylenetetrazole 15-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 8542328-6 1995 Haloperidol, chlorpromazine, raclopride and risperidone all significantly increased Fos expression in the medial and lateral striatum. Haloperidol 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 8542328-6 1995 Haloperidol, chlorpromazine, raclopride and risperidone all significantly increased Fos expression in the medial and lateral striatum. Chlorpromazine 13-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 8542328-6 1995 Haloperidol, chlorpromazine, raclopride and risperidone all significantly increased Fos expression in the medial and lateral striatum. Raclopride 29-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 8542328-6 1995 Haloperidol, chlorpromazine, raclopride and risperidone all significantly increased Fos expression in the medial and lateral striatum. Risperidone 44-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 8542328-8 1995 Haloperidol, thioridazine and risperidone also markedly increased Fos expression in the lateral septum. Haloperidol 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 8542328-8 1995 Haloperidol, thioridazine and risperidone also markedly increased Fos expression in the lateral septum. Thioridazine 13-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 8542328-8 1995 Haloperidol, thioridazine and risperidone also markedly increased Fos expression in the lateral septum. Risperidone 30-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 8542328-9 1995 Distinguishing it from the other neuroleptics, clozapine did not increase Fos expression in the lateral striatum, but induced a significant increase in Fos expression in the prefrontal cortex. Clozapine 47-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-155 8564192-19 1995 An indicator of neuronal activation, c-Fos-like immunoreactivity, was detected in the ipsilateral, but not in the contralateral frontoparietal cortex 2 h after KCl application. Potassium Chloride 160-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 8564192-27 1995 These findings suggest that the inhibitory effects of KB-2796 and flunarizine on the cortical hypoperfusion and expression of c-Fos-like immunoreactivity induced by spreading depression are mediated via the effects of Ca(2+)-entry blockade, which may include an increase in cerebral blood flow and the prevention of excessive Ca2+ influx into brain cells. lomerizine 54-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 8564192-27 1995 These findings suggest that the inhibitory effects of KB-2796 and flunarizine on the cortical hypoperfusion and expression of c-Fos-like immunoreactivity induced by spreading depression are mediated via the effects of Ca(2+)-entry blockade, which may include an increase in cerebral blood flow and the prevention of excessive Ca2+ influx into brain cells. Flunarizine 66-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 7494448-6 1995 Nicotine administration by injection increased the phosphorylation of CREB and induced c-Fos protein without affecting members of the jun family. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 7634414-2 1995 In the present study we have investigated whether topically administered c-fos antisense oligodeoxynucleotides (ODNs) inhibit c-fos activation in the UV-exposed rat skin and thereby modulate the delayed increase in cellular proliferative activity. Oligodeoxyribonucleotides 89-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 7634414-2 1995 In the present study we have investigated whether topically administered c-fos antisense oligodeoxynucleotides (ODNs) inhibit c-fos activation in the UV-exposed rat skin and thereby modulate the delayed increase in cellular proliferative activity. Oligodeoxyribonucleotides 112-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 7634414-2 1995 In the present study we have investigated whether topically administered c-fos antisense oligodeoxynucleotides (ODNs) inhibit c-fos activation in the UV-exposed rat skin and thereby modulate the delayed increase in cellular proliferative activity. Oligodeoxyribonucleotides 112-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 7581259-5 1995 This ANG II-induced c-fos mRNA expression was totally inhibited by icv pretreatment with the ANG II-AT1 receptor antagonist, losartan. Losartan 125-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 7671799-3 1995 Here we show, that GABA and the GABAA receptor agonist muscimol induce c-Fos immunoreactivity and increase BDNF mRNA expression in embryonic hippocampal neurons cultured for 5 days. gamma-Aminobutyric Acid 19-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 7671799-3 1995 Here we show, that GABA and the GABAA receptor agonist muscimol induce c-Fos immunoreactivity and increase BDNF mRNA expression in embryonic hippocampal neurons cultured for 5 days. Muscimol 55-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 7671799-6 1995 The increase in c-Fos immunoreactivity and BDNF mRNA levels by GABA were dependent upon the activation of voltage-gated Ca2+ channels, as shown using the L-type specific Ca2+ channel blocker nifedipine. gamma-Aminobutyric Acid 63-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 7671799-6 1995 The increase in c-Fos immunoreactivity and BDNF mRNA levels by GABA were dependent upon the activation of voltage-gated Ca2+ channels, as shown using the L-type specific Ca2+ channel blocker nifedipine. Nifedipine 191-201 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 7671799-7 1995 The differential regulation of c-fos and BDNF expression by GABA and muscimol in developing and mature hippocampal neurons is due to a switch in the ability of GABAA receptors to activate voltage-gated Ca2+ channels. gamma-Aminobutyric Acid 60-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 7671799-7 1995 The differential regulation of c-fos and BDNF expression by GABA and muscimol in developing and mature hippocampal neurons is due to a switch in the ability of GABAA receptors to activate voltage-gated Ca2+ channels. Muscimol 69-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 7628395-9 1995 These associative data were strengthened by our observation that in paradigms in which labor was induced prematurely by ovariectomy or blocked by treatment with progesterone, changes in c-fos expression were closely matched by changes in Cx-43 mRNA. Progesterone 161-173 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 186-191 7635978-0 1995 Linoleic acid and its metabolites, hydroperoxyoctadecadienoic acids, stimulate c-Fos, c-Jun, and c-Myc mRNA expression, mitogen-activated protein kinase activation, and growth in rat aortic smooth muscle cells. Linoleic Acid 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 7635978-0 1995 Linoleic acid and its metabolites, hydroperoxyoctadecadienoic acids, stimulate c-Fos, c-Jun, and c-Myc mRNA expression, mitogen-activated protein kinase activation, and growth in rat aortic smooth muscle cells. hydroperoxyoctadecadienoic acids 35-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 7635978-4 1995 Linoleic acid induces DNA synthesis, c-fos, c-jun, and c-myc mRNA expression and MAPK activation in VSMC. Linoleic Acid 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 7476995-7 1995 AP-1, cAMP response element, and glucocorticoid response element motifs are required for full cooperative activation by either c-Jun or c-Jun/c-Fos and glucocorticoids. Cyclic AMP 6-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-147 7675199-0 1995 Cholecystokinin B receptor antagonism enhances the ability of a low dose of morphine to reduce c-Fos expression in the spinal cord of the rat. Morphine 76-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 7675199-1 1995 Three hours after intraplantar carrageenin (6 mg/150 microliters) Fos-like immunoreactivity was predominantly observed in the superficial and deep laminae of the L4-L5 segments of the dorsal horn of the spinal cord in the rat. Carrageenan 31-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 7675199-5 1995 However, pre-administration of a higher dose of morphine (3 mg/kg) significantly reduced the total number of Fos-like immunoreactive neurons (28 +/- 8% reduction, P < 0.001, as compared with control carrageenin Fos-like immunoreactive expression), with this effect being equally divided between the superficial and deep laminae (29 +/- 5 and 29 +/- 6% reduction, respectively, P < 0.001, as compared with control carrageenin Fos-like immunoreactive expression, for both). Morphine 48-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-112 7675199-5 1995 However, pre-administration of a higher dose of morphine (3 mg/kg) significantly reduced the total number of Fos-like immunoreactive neurons (28 +/- 8% reduction, P < 0.001, as compared with control carrageenin Fos-like immunoreactive expression), with this effect being equally divided between the superficial and deep laminae (29 +/- 5 and 29 +/- 6% reduction, respectively, P < 0.001, as compared with control carrageenin Fos-like immunoreactive expression, for both). Morphine 48-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 214-217 7675199-5 1995 However, pre-administration of a higher dose of morphine (3 mg/kg) significantly reduced the total number of Fos-like immunoreactive neurons (28 +/- 8% reduction, P < 0.001, as compared with control carrageenin Fos-like immunoreactive expression), with this effect being equally divided between the superficial and deep laminae (29 +/- 5 and 29 +/- 6% reduction, respectively, P < 0.001, as compared with control carrageenin Fos-like immunoreactive expression, for both). Morphine 48-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 214-217 8539319-9 1995 Rats conditioned with cocaine, but not U-50,488, showed increased Fos activity in the anterior cingulate cortex, piriform cortex, lateral septal area, and olfactory tubercles. Cocaine 22-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 8542301-7 1995 Intraperitoneal administration of ondansetron or perivagal capsaicin treatment significantly reduced the duration of MMC disruption and attenuated markedly c-fos staining in the 3 brain sites. Ondansetron 34-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 8542301-7 1995 Intraperitoneal administration of ondansetron or perivagal capsaicin treatment significantly reduced the duration of MMC disruption and attenuated markedly c-fos staining in the 3 brain sites. Capsaicin 59-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 8542301-9 1995 Blockade of both c-fos expression and MMC disruption by systemic ondansetron and by perivagal capsaicin indicates that some brainstem nuclei are involved in digestive disturbances after intestinal anaphylaxis, and reflects an involvement of peripheral 5-HT3 receptors on vagal afferents. Ondansetron 65-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 8542301-9 1995 Blockade of both c-fos expression and MMC disruption by systemic ondansetron and by perivagal capsaicin indicates that some brainstem nuclei are involved in digestive disturbances after intestinal anaphylaxis, and reflects an involvement of peripheral 5-HT3 receptors on vagal afferents. Capsaicin 94-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 8542301-10 1995 The reduction of c-fos staining in NTS as well as in LPB and PVN after perivagal capsaicin suggests that the NTS is the primary relay in the activation of the central nervous system during intestinal allergic challenge. Capsaicin 81-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 8539319-10 1995 When considered together, these results suggest that U-50,488 was effective at blocking the unconditioned and conditioned effects of cocaine, as well as cocaine-induced neuronal activity (as measured by Fos induction). Cocaine 153-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 203-206 7482290-3 1995 Fos immunoreactivity induced by d-amphetamine (5 mg/kg) or apomorphine (5 mg/kg) was quantified in dorsolateral and ventrolateral regions of caudate-putamen (CPu) in rats still demonstrating sensory neglect (5 days postsurgery) and in rats recovered from sensory neglect produced by AGm ablation (29+ days postsurgery). Dextroamphetamine 32-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 7482290-3 1995 Fos immunoreactivity induced by d-amphetamine (5 mg/kg) or apomorphine (5 mg/kg) was quantified in dorsolateral and ventrolateral regions of caudate-putamen (CPu) in rats still demonstrating sensory neglect (5 days postsurgery) and in rats recovered from sensory neglect produced by AGm ablation (29+ days postsurgery). Apomorphine 59-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 7482290-0 1995 Time-dependent changes in dopamine agonist-induced striatal Fos immunoreactivity are related to sensory neglect and its recovery after unilateral prefrontal cortex injury. Dopamine 26-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 7482290-5 1995 In rats demonstrating sensory neglect, d-amphetamine or apomorphine induction of Fos in the ipsilateral CPu was reduced by about 40% compared to the contralateral CPu or to comparable readings in unlesioned controls. Dextroamphetamine 39-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-84 7482290-5 1995 In rats demonstrating sensory neglect, d-amphetamine or apomorphine induction of Fos in the ipsilateral CPu was reduced by about 40% compared to the contralateral CPu or to comparable readings in unlesioned controls. Apomorphine 56-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-84 7478217-6 1995 This suggests that the increase in striatal Fos levels observed following dopamine depletion may occur as a result of elevated cytoplasmic calcium levels in the striatal cells. Dopamine 74-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 7544033-4 1995 In response to terbutaline, a beta 2-selective agonist, marked stimulation of c-fos was demonstrated in the liver, which contains beta 2-receptors, on Gestational Day 20, as well as on Postnatal Days 1 and 8. Terbutaline 15-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 7544033-5 1995 In the heart, which contains predominantly beta 1-receptors, isoproterenol (a non-subtype-selective beta-agonist) was more effective that terbutaline, indicating that either receptor subtype can elicit stimulation of c-fos. Isoproterenol 61-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 217-222 7544033-5 1995 In the heart, which contains predominantly beta 1-receptors, isoproterenol (a non-subtype-selective beta-agonist) was more effective that terbutaline, indicating that either receptor subtype can elicit stimulation of c-fos. Terbutaline 138-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 217-222 7544033-8 1995 The intracellular signaling cascade from beta-receptor to c-fos expression may thus provide one of the basic cellular mechanisms for trophic control of differentiation by biogenic amines, and for the teratologies associated with beta-adrenergic agonist therapy. Amines 180-186 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 7478238-1 1995 This study was performed to determine whether c-fos immunoreactivity (IR) induced in medulla oblongata by pentylenetetrazole seizures is a consequence of seizure-associated blood pressure elevation and activation of baroreceptor afferent pathways, or occurs independently of hypertension. Pentylenetetrazole 106-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 7478238-2 1995 Immunohistochemical study of sections of medulla oblongata revealed that seizures are followed by induction of c-fos IR in nucleus tractus solitarius (NTS), dorsal motor nucleus of the vagus (DMN 10), and ventrolateral medulla (VLM), while there is negligible c-fos IR after saline sham injections. Sodium Chloride 275-281 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 7478238-5 1995 phenylephrine also resulted in induction of c-fos IR in NTS. Phenylephrine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 7478217-0 1995 Elevated striatal Fos immunoreactivity following 6-hydroxydopamine lesioning of the rat is mediated by excitatory amino acid transmission. Oxidopamine 49-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 7478217-0 1995 Elevated striatal Fos immunoreactivity following 6-hydroxydopamine lesioning of the rat is mediated by excitatory amino acid transmission. Excitatory Amino Acids 103-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 7478217-3 1995 To test the hypothesis that elevated N-methyl-D-aspartic acid (NMDA)-mediated corticostriatal transmission may underlie the increase in striatal Fos levels upon dopamine depletion, rats were unilaterally 6-hydroxydopamine lesioned under anaesthesia induced by either barbiturate or the NMDA antagonist, ketamine. N-Methylaspartate 37-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-148 7478217-3 1995 To test the hypothesis that elevated N-methyl-D-aspartic acid (NMDA)-mediated corticostriatal transmission may underlie the increase in striatal Fos levels upon dopamine depletion, rats were unilaterally 6-hydroxydopamine lesioned under anaesthesia induced by either barbiturate or the NMDA antagonist, ketamine. N-Methylaspartate 63-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-148 7478217-3 1995 To test the hypothesis that elevated N-methyl-D-aspartic acid (NMDA)-mediated corticostriatal transmission may underlie the increase in striatal Fos levels upon dopamine depletion, rats were unilaterally 6-hydroxydopamine lesioned under anaesthesia induced by either barbiturate or the NMDA antagonist, ketamine. Dopamine 161-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-148 7478217-5 1995 The results demonstrate that dopamine depletion following 6-hydroxydopamine lesioning can result in elevated striatal Fos levels which can be attenuated by contiguous treatment with an NMDA antagonist. Dopamine 29-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 7478217-5 1995 The results demonstrate that dopamine depletion following 6-hydroxydopamine lesioning can result in elevated striatal Fos levels which can be attenuated by contiguous treatment with an NMDA antagonist. Oxidopamine 58-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 7478217-5 1995 The results demonstrate that dopamine depletion following 6-hydroxydopamine lesioning can result in elevated striatal Fos levels which can be attenuated by contiguous treatment with an NMDA antagonist. N-Methylaspartate 185-189 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 7488730-0 1995 Carrageenan oedema and spinal Fos-LI neurones are reduced by piroxicam in the rat. Piroxicam 61-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 7488730-1 1995 Intraplantar carrageenan induced a peripheral oedema and c-Fos like immunoreactivity (Fos-LI) in L4-L5 segments in the dorsal horn (DH) of the rat spinal cord. Carrageenan 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 7488730-1 1995 Intraplantar carrageenan induced a peripheral oedema and c-Fos like immunoreactivity (Fos-LI) in L4-L5 segments in the dorsal horn (DH) of the rat spinal cord. Carrageenan 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 7488730-2 1995 The total number of carrageenan evoked Fos-LI neurones was 153 +/- 10 per section, with a predominant localization in the superficial and deep laminae of the DH (46 +/- 4% and 32 +/- 2%, respectively). Carrageenan 20-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 7488730-5 1995 The effects of piroxicam on the number of spinal Fos-LI neurones and on ankle oedema were positively correlated. Piroxicam 15-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 7478217-6 1995 This suggests that the increase in striatal Fos levels observed following dopamine depletion may occur as a result of elevated cytoplasmic calcium levels in the striatal cells. Calcium 139-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 7478218-0 1995 The effect of clozapine on Fos protein immunoreactivity in the rat forebrain is not mimicked by the addition of alpha 1-adrenergic or 5HT2 receptor blockade to haloperidol. Clozapine 14-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 7478218-1 1995 The involvement of alpha 1-adrenergic and 5HT2-receptor blockade in the induction of Fos protein produced by the "atypical" neuroleptic clozapine was investigated in the rat forebrain. Clozapine 136-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 7478218-3 1995 Clozapine (20 mg/kg) induced a significantly higher Fos protein immunoreactivity response in the medial prefrontal cortex and a significantly lower response in the dorsolateral striatum compared to the effect of haloperidol (1 mg/kg). Clozapine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 7478176-1 1995 Rats were either depleted of sodium by treatment with a diuretic or were made hypovolemic by injection of polyethylene glycol (PEG) and their brains were subsequently examined for induction of Fos-like immunoreactivity (FLI). Polyethylene Glycols 127-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-196 7583228-0 1995 Cholecystokinin octapeptide and the D2 antagonist raclopride induce Fos-like immunoreactivity in the shell part of the rat nucleus accumbens via different mechanisms. Raclopride 50-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 7583228-1 1995 Induction of neuronal Fos-like immunoreactivity (IR) in the rat brain by cholecystokinin octapeptide (CCK-8) and the dopamine (DA) D2 receptor antagonist raclopride was demonstrated. Dopamine 117-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-25 7583228-1 1995 Induction of neuronal Fos-like immunoreactivity (IR) in the rat brain by cholecystokinin octapeptide (CCK-8) and the dopamine (DA) D2 receptor antagonist raclopride was demonstrated. Raclopride 154-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-25 7583228-4 1995 alone increased in a dose-dependent way the Fos-like ir profiles in the shell part of the rat nucleus accumbens (AcbSh). acbsh 113-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 7583228-5 1995 Combined treatment with CCK-8 (0.1 nmol/rat) and raclopride (0.5 mg/kg) caused significant additive increases in the Fos-like ir profiles in the AcbSh. Raclopride 49-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 7788922-13 1995 Treatment with TCV-116 significantly inhibited the induction of mRNAs for c-fos, c-jun, Egr-1, ODC, and fibronectin in injured artery, whereas the increase in TGF-beta 1 gene expression after injury was not prevented by TCV-116. candesartan cilexetil 15-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 7673484-2 1995 In pentobarbital anesthetized rats, the expression of the c-FOS protein was detected by immunocytochemistry and was used as a marker of neuronal activity. Pentobarbital 3-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 7789326-0 1995 Cellular localization of estradiol-induced c-fos messenger ribonucleic acid in the rat uterus: c-fos expression and uterine cell proliferation do not correlate strictly. Estradiol 25-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 7789326-0 1995 Cellular localization of estradiol-induced c-fos messenger ribonucleic acid in the rat uterus: c-fos expression and uterine cell proliferation do not correlate strictly. Estradiol 25-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 7789326-6 1995 In both immature and mature castrated rats at 3 h after 17 beta-estradiol administration, c-fos expression was detected primarily in uterine luminal and glandular epithelia. Estradiol 56-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 7789326-7 1995 Expression of c-fos returned to baseline levels by 24 h post 17 beta-estradiol treatment. Estradiol 61-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 7789326-10 1995 Treatment of adult rats with a single injection of 16 alpha-estradiol, a short-acting, nonmitogenic estrogen, induced c-fos primarily in the uterine glandular epithelia. estradiol-16 51-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 7615792-4 1995 In addition, we determined the effects of adrenalectomy and dexamethasone administration on c-fos and CRF gene expression in the PVN. Dexamethasone 60-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 8948900-0 1995 [Effect of adrenaline on food consumption and C-fos expression in the central nervous system]. Epinephrine 11-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 7623110-2 1995 In rats that performed a well-learned nose-poke response for water reward, c-fos mRNA levels were broadly increased, relative to values in home cage-control rats, in visual cortex, superior colliculus, olfactory bulb, and, to comparable levels, regions CA3 and CA1 of hippocampus; hybridization was not increased in the dentate gyrus. Water 61-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 7623110-3 1995 In rats first trained on the nose-poke behavior and then required to discriminate between two odors for water reward, the increase in c-fos mRNA was generally not as great and was more regionally differentiated. Water 104-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-139 7623133-3 1995 In conscious 21 d pregnant rats naloxone increased the number of neurons expressing Fos (an indicator of neuronal activity) in the supraoptic nucleus (SON) but had no effect on 16 d pregnant or virgin rats. Naloxone 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 7477894-0 1995 Regional suppression by lesions in the anterior third ventricle of c-fos expression induced by either angiotensin II or hypertonic saline. Sodium Chloride 131-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 7589783-6 1995 Under identical conditions, TPA and serum rapidly induce c-fos transcription in RENE1 cells, indicating that the lack of inducibility by estradiol is not due to a general inhibitor of transcription of these genes. Tetradecanoylphorbol Acetate 28-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 7552308-0 1995 Systemic nitroglycerin induces Fos immunoreactivity in brainstem and forebrain structures of the rat. Nitroglycerin 9-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 7552308-3 1995 In this study, we evaluated the distribution and intensity of Fos immunoreactivity in rat brain nuclei following the systemic administration of nitroglycerin. Nitroglycerin 144-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 7608202-5 1995 An oligonucleotide representing the AP-1 recognition sequence also blocked the NFAT DNA binding activity, as did a combination of anti-Fos and anti-Jun antibodies. Oligonucleotides 3-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-138 7477908-0 1995 Adenosine A2 receptors stimulate c-fos expression in striatal neurons of 6-hydroxydopamine-lesioned rats. Oxidopamine 73-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 7477908-1 1995 The induction of the early-gene c-fos after administration of the adenosine A2a receptor agonist CGS 21680, was studied in the striatum of normal rats or in rats with a unilateral 6-hydroxydopamine lesion of the dopaminergic nigrostriatal neurons. cysteinylglycine 97-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 7477908-2 1995 CGS 21680 (2.25 mg/kg) induces c-fos expression in the 6-hydroxydopamine-lesioned striatum, while up to 40 mg/kg fails to induce c-fos in the intact striatum or in the striatum of normal rats. Oxidopamine 55-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 7477908-3 1995 Blockade of muscarine receptors by scopolamine (5 mg/kg) partially prevents, and stimulation of dopamine D2 receptors by quinpirole (0.5 mg/kg) completely reverses, CGS 21680-induced c-fos expression in the 6-hydroxydopamine-lesioned striatum. Quinpirole 121-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 183-188 7477908-3 1995 Blockade of muscarine receptors by scopolamine (5 mg/kg) partially prevents, and stimulation of dopamine D2 receptors by quinpirole (0.5 mg/kg) completely reverses, CGS 21680-induced c-fos expression in the 6-hydroxydopamine-lesioned striatum. cysteinylglycine 165-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 183-188 7477908-3 1995 Blockade of muscarine receptors by scopolamine (5 mg/kg) partially prevents, and stimulation of dopamine D2 receptors by quinpirole (0.5 mg/kg) completely reverses, CGS 21680-induced c-fos expression in the 6-hydroxydopamine-lesioned striatum. Oxidopamine 207-224 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 183-188 7477908-4 1995 In turn, CGS 21680 partially reverses c-fos expression induced by quinpirole in the lesioned globus pallidus. Quinpirole 66-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 7477908-6 1995 The results suggest that CGS 21680 induces c-fos expression in the striatum through direct and indirect mechanisms related to the ability of A2a receptors to stimulate cyclic AMP formation or acetylcholine release which in turn would activate c-fos through muscarinic receptors. Cyclic AMP 168-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 7655348-3 1995 Kainic acid causes an increase in metallothionein-I and heme oxygenase-I mRNAs, as well as an increase in c-fos, heat shock protein-70, and interleukin-1 beta mRNAs. Kainic Acid 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 7675181-1 1995 The expression of the proto-oncogene c-fos was used as a cellular marker of spinal cord neurons activated by microinjection of kainic acid into the medullary nucleus raphe magnus of awake and drug-free Sprague-Dawley rats. Kainic Acid 127-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-42 7552347-0 1995 Distribution of Fos-positive neurons in cortical and subcortical structures after picrotoxin-induced convulsions varies with seizure type. Picrotoxin 82-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 7642805-2 1995 Fos-like immunoreactivity (Fos-LI) in the caudal brainstem was visualized 2 hours after unilateral injection of the small-fiber-specific excitant/inflammatory irritant mustard oil into the TMJ region. mustard oil 168-179 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 7642805-2 1995 Fos-like immunoreactivity (Fos-LI) in the caudal brainstem was visualized 2 hours after unilateral injection of the small-fiber-specific excitant/inflammatory irritant mustard oil into the TMJ region. mustard oil 168-179 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 7642805-8 1995 Compared to TMJ mustard oil injection, mineral oil injection produced less Fos-LI at all rostrocaudal levels, whereas subcutaneous mustard oil injection produced less Fos-LI in caudal subnucleus caudalis but similar amounts in the cervical dorsal horn. Mineral Oil 39-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 7615792-8 1995 Adrenalectomy augmented and dexamethasone pretreatment inhibited c-fos mRNA, CRF hnRNA, and mRNA induction after stress. Dexamethasone 28-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 7790908-0 1995 Kainic acid-induced neuronal death is associated with DNA damage and a unique immediate-early gene response in c-fos-lacZ transgenic rats. Kainic Acid 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 7667349-0 1995 Chronic desipramine alters stress-induced behaviors and regional expression of the immediate early gene, c-fos. Desipramine 8-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 7649226-7 1995 Our analysis indicates that c-fos expression increases soon after quinolinic acid injection, is widespread in rat brain, but is effectively absent by 24 h postinjection. Quinolinic Acid 66-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 7791123-0 1995 Rotation and striatal c-fos expression after repeated, daily treatment with selective dopamine receptor agonists and levodopa. Levodopa 117-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 7651620-0 1995 Acute injection of adrenal steroids reduces cornea-evoked expression of c-fos within the spinal trigeminal nucleus of adrenalectomized rats. Steroids 27-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 7651620-1 1995 The influence of transient increases in adrenal steroid hormones on the number of Fos-positive neurons after nociceptor activation was assessed in adrenalectomized rats. Steroids 48-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-85 7651620-3 1995 Adrenalectomized rats displayed an enhanced number of Fos-positive neurons within the caudal-most portions of trigeminal subnucleus caudalis compared to that seen in adrenal-intact animals, an effect reversed by a single acute injection of corticosterone (1 mg/kg, i.p.) Corticosterone 240-254 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 7651620-5 1995 Acute injection of the selective mineralocorticoid receptor agonist, aldosterone, or the selective glucocorticoid receptor agonist, RU28362, also reduced the number of Fos-positive neurons. Aldosterone 69-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-171 7651620-5 1995 Acute injection of the selective mineralocorticoid receptor agonist, aldosterone, or the selective glucocorticoid receptor agonist, RU28362, also reduced the number of Fos-positive neurons. 11,17-dihydroxy-6-methyl-17-(1-propynyl)androsta-1,4,6-triene-3-one 132-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-171 7651620-6 1995 Aldosterone and RU28362 had an additive effect on Fos when given concurrently. 11,17-dihydroxy-6-methyl-17-(1-propynyl)androsta-1,4,6-triene-3-one 16-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 7651620-8 1995 Acute administration of adrenal steroids to adrenalectomized rats greatly attenuated the number of Fos-positive neurons seen after corneal stimulation within select portions of trigeminal subnucleus caudalis. Steroids 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 7651620-10 1995 These results are consistent with the hypothesis that transient increases in adrenal steroids, such as occur after injury, are sufficient to modify the production of Fos protein in central neurons that process nociceptive information. Steroids 85-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-169 7667349-1 1995 This experiment examined the effects of acute or chronic administration of the antidepressant drug desipramine on conditioned stress-induced behaviors and regional c-fos expression in the brain. Desipramine 99-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-169 7667349-7 1995 Similarly, Fos immunohistochemistry revealed that the chronic desipramine group showing positive behavioral effects was the only group in which there were significant reductions in the number of stress-induced Fos-positive neurons in five of 60 structures surveyed. Desipramine 62-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 7667349-7 1995 Similarly, Fos immunohistochemistry revealed that the chronic desipramine group showing positive behavioral effects was the only group in which there were significant reductions in the number of stress-induced Fos-positive neurons in five of 60 structures surveyed. Desipramine 62-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 210-213 7632891-0 1995 Endogenous histamine induces c-fos expression within paraventricular and supraoptic nuclei. Histamine 11-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 8612146-0 1995 Induction of c-fos and reduction of dynorphin in dentate granule cells of a rat model of epilepsy produced by systemic administration of kainic acid: an immunohistochemical study. Kainic Acid 137-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 7659789-0 1995 Pregastric and postabsorptive inhibitory effects of water on angiotensin-induced Fos in rat brain. Water 52-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-84 7659789-2 1995 It has been reported that the Fos-like immunoreactivity (FLI; a marker of cell activation) that is induced in PVN and SON by exogenous Ang II is prevented by allowing rats to ingest water after injection. Water 182-187 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 7750495-6 1995 The double labeled in situ hybridization results demonstrated that TRH and c-fos/c-jun are coexpressed in anterior pituitary cells and that DEX (10(-8) M) enhanced the cell intensity for TRH and c-fos/c-jun. Dexamethasone 140-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 7750495-6 1995 The double labeled in situ hybridization results demonstrated that TRH and c-fos/c-jun are coexpressed in anterior pituitary cells and that DEX (10(-8) M) enhanced the cell intensity for TRH and c-fos/c-jun. Dexamethasone 140-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-200 7750495-8 1995 The Northern blot analysis also showed that DEX increased the messenger RNA level of TRH 5.1-fold (n = 4; P < 0.01), that of c-fos 1.8-fold (n = 5; P < 0.01), and that of c-jun 4.2-fold (n = 4; P < 0.01). Dexamethasone 44-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-133 7750495-9 1995 The nuclear run-on analysis indicated that DEX increased the nuclear transcription activity of TRH 3.3-fold, that of c-jun 3.2-fold, and that of c-fos 3-fold (n = 3; P < 0.01) vs. the control value. Dexamethasone 43-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-150 7750495-10 1995 The coexpression of TRH and c-fos/c-jun in anterior pituitary cells as well as the enhancement of transcription after DEX treatment raise the possibility that c-fos/c-jun could mediate the effect of GC on TRH gene transcriptional activity. Dexamethasone 118-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-164 7632891-1 1995 Thioperamide, an H3-receptor antagonist that enhances endogenous histamine release, induced c-fos mRNA expression and Fos-like immunoreactivity in magnocellular neurones of rat supraoptic and paraventricular nuclei. thioperamide 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 7632891-1 1995 Thioperamide, an H3-receptor antagonist that enhances endogenous histamine release, induced c-fos mRNA expression and Fos-like immunoreactivity in magnocellular neurones of rat supraoptic and paraventricular nuclei. thioperamide 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 7648263-0 1995 Sodium depletion induces Fos immunoreactivity in circumventricular organs of the lamina terminalis. Sodium 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 7648263-1 1995 Acute sodium depletion by peritoneal dialysis (PD) induces c-fos expression in the subfornical organ (SFO) and organum vasculosum laminae terminalis (OVLT), in conscious rats. Sodium 6-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 7648263-4 1995 When rats were allowed to drink sodium salt (1.8% NaCl) 24 h after PD, there was a marked reversion of the c-fos expression in the OVLT and a comparatively smaller effect in the SFO. sodium salt 32-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 7648263-4 1995 When rats were allowed to drink sodium salt (1.8% NaCl) 24 h after PD, there was a marked reversion of the c-fos expression in the OVLT and a comparatively smaller effect in the SFO. Sodium Chloride 50-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 7648263-5 1995 Intracerebroventricular infusion of hypertonic CSF (170 mM NaCl) from 30 min before and during 4 h after PD, significantly inhibited the c-fos expression in both nuclei. Sodium Chloride 59-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 7648263-6 1995 These results demonstrate that an acute body sodium deficit induces c-fos activity in SFO and OVLT neurons, indicating the special role of these structures in sodium balance regulation. Sodium 45-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 7648263-6 1995 These results demonstrate that an acute body sodium deficit induces c-fos activity in SFO and OVLT neurons, indicating the special role of these structures in sodium balance regulation. Sodium 159-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 7648263-7 1995 They also show that the sodium-depletion-induced production of Fos in neurons of the lamina terminalis can be modulated by central or systemic reposition of sodium. Sodium 24-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 7648263-7 1995 They also show that the sodium-depletion-induced production of Fos in neurons of the lamina terminalis can be modulated by central or systemic reposition of sodium. Sodium 157-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 7648269-0 1995 Activation and desensitization of Fos immunoreactivity in the rat brain following ethanol administration. Ethanol 82-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 7648269-3 1995 Following intraperitoneal injection of ethanol (16% w/v), Fos immunoreactivity was induced in several rat brain areas including the bed nucleus of the stria terminalis, paraventricular hypothalamic nucleus, the central nucleus of amygdala, Edinger-Westphal nucleus, locus coeruleus nucleus and parabrachial nucleus. Ethanol 39-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 7648269-5 1995 Fos immunoreactivity in the supraoptic nucleus appeared only when a higher concentration of ethanol was injected. Ethanol 92-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 7648269-6 1995 Repeated administration of ethanol twice daily for 17 or 24 days resulted in a desensitization of Fos immunoreactivity in these nuclei. Ethanol 27-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 7648269-7 1995 These data suggest that induction of Fos immunoreactivity can be used to determine the sites at which ethanol acts on the brain, and may provide important information about the mechanisms underlying the tolerance and physical dependence of alcohol usage. Ethanol 102-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 7648269-7 1995 These data suggest that induction of Fos immunoreactivity can be used to determine the sites at which ethanol acts on the brain, and may provide important information about the mechanisms underlying the tolerance and physical dependence of alcohol usage. Alcohols 240-247 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 7572198-0 1995 Effect of different xanthines and phosphodiesterase inhibitors on c-fos expression in rat striatum. Xanthines 20-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 7572198-1 1995 It has previously been shown that caffeine, in a dose-dependent manner, increases the expression of the protooncogene c-fos in the rat brain, predominantly in the caudate-putamen and tuberculum olfactorium. Caffeine 34-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 7572198-2 1995 In this study we examined the effect of related xanthines and of selective phosphodiesterase inhibitors on c-fos expression. Xanthines 48-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 7572198-5 1995 Thus, c-fos is induced in rat striatum following administration of caffeine and other xanthines that (provided they enter the brain) block adenosine receptors, suggesting an involvement of central adenosine receptors. Caffeine 67-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 6-11 7743507-0 1995 Induction of c-fos and c-jun proto-oncogene expression by asbestos is ameliorated by N-acetyl-L-cysteine in mesothelial cells. Asbestos 58-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 7743507-0 1995 Induction of c-fos and c-jun proto-oncogene expression by asbestos is ameliorated by N-acetyl-L-cysteine in mesothelial cells. Acetylcysteine 85-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 7743507-1 1995 Asbestos fibers cause dose-dependent, persistent increases in mRNA levels of c-jun and c-fos proto-oncogenes in rat pleural mesothelial (RPM) cells, the progenitor cells of asbestos-induced mesothelioma (N. Heintz, Y. M. W. Janssen, and B. T. Mossman. Asbestos 173-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 7743507-7 1995 Here we report that addition of N-acetyl-L-cysteine decreases asbestos-mediated induction of c-fos and c-jun mRNA levels in a dose-dependent fashion. Acetylcysteine 32-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 7743507-7 1995 Here we report that addition of N-acetyl-L-cysteine decreases asbestos-mediated induction of c-fos and c-jun mRNA levels in a dose-dependent fashion. Asbestos 62-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 7743507-9 1995 Pretreatment of cells with buthionine sulfoximine, an agent which diminishes glutathione pools, increases the magnitude of induction of c-fos and c-jun mRNA by asbestos. Buthionine Sulfoximine 27-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 7743507-9 1995 Pretreatment of cells with buthionine sulfoximine, an agent which diminishes glutathione pools, increases the magnitude of induction of c-fos and c-jun mRNA by asbestos. Glutathione 77-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 7743507-14 1995 However, intracellular thiol levels appear to influence the expression of c-fos and c-jun, suggesting a redox-sensitive component in the signaling cascade which modulates gene expression of c-fos and c-jun by asbestos. Sulfhydryl Compounds 23-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 7743507-14 1995 However, intracellular thiol levels appear to influence the expression of c-fos and c-jun, suggesting a redox-sensitive component in the signaling cascade which modulates gene expression of c-fos and c-jun by asbestos. Sulfhydryl Compounds 23-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 190-195 7538563-9 1995 Amphetamine-induced striatal Fos expression was normalized in the caudate-putamen ipsilateral to the intranigral VM grafts, showing hyperexpression in some areas of the striatum, and the apomorphine-induced Fos expression seen in the 6-OHDA-lesioned animals was completely reversed on the grafted side. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 7538563-9 1995 Amphetamine-induced striatal Fos expression was normalized in the caudate-putamen ipsilateral to the intranigral VM grafts, showing hyperexpression in some areas of the striatum, and the apomorphine-induced Fos expression seen in the 6-OHDA-lesioned animals was completely reversed on the grafted side. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 207-210 7538563-9 1995 Amphetamine-induced striatal Fos expression was normalized in the caudate-putamen ipsilateral to the intranigral VM grafts, showing hyperexpression in some areas of the striatum, and the apomorphine-induced Fos expression seen in the 6-OHDA-lesioned animals was completely reversed on the grafted side. Apomorphine 187-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 7538563-9 1995 Amphetamine-induced striatal Fos expression was normalized in the caudate-putamen ipsilateral to the intranigral VM grafts, showing hyperexpression in some areas of the striatum, and the apomorphine-induced Fos expression seen in the 6-OHDA-lesioned animals was completely reversed on the grafted side. Oxidopamine 234-240 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 7537689-5 1995 Nearly all neurons expressed c-Fos after the treatment with colchicine. Colchicine 60-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 7751931-1 1995 The effect of a single injection of caffeine on the expression of c-fos, c-jun, junB, and junD, on activator protein 1 (AP-1) and on the levels of preproenkephalin mRNA in rat striatum was studied. Caffeine 36-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 7751931-4 1995 By using in situ hybridization of adjacent sections we found a rapid, transient, and dose-dependent increase of c-fos, c-jun, and junB by caffeine in striatum, especially in the lateral part. Caffeine 138-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 7751931-12 1995 The data show that a single dose of caffeine induces a temporally and spatially characteristic pattern of c-fos, c-jun, and junB induction, followed by changes in AP-1 and preproenkephalin mRNA. Caffeine 36-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 7637264-8 1995 Gentamicin induced c-fos mRNA expression in quiescent mesangial cells. Gentamicins 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 7566485-2 1995 Fos was detected in many magno- and parvocellular NADPH-D positive neurons in response to haemorrhage or drug-evoked hypotension using i.v. NADP 50-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 7566487-7 1995 The induction of c-fos mRNA by kainate injection was most sensitive to dexamethasone (2 mg/kg), whereas a higher dose (30 mg/kg) was required to attenuate the induction of zif-268 mRNA. Kainic Acid 31-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 7566487-7 1995 The induction of c-fos mRNA by kainate injection was most sensitive to dexamethasone (2 mg/kg), whereas a higher dose (30 mg/kg) was required to attenuate the induction of zif-268 mRNA. Dexamethasone 71-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 7566487-8 1995 These results show that a time-dependent and co-ordinated induction of c-fos, c-jun, jun-B and zif-268 mRNA in cerebral cortex occurs in response to the persistent excitation caused by excitotoxin injection which is mediated by glutamate and shows a differential sensitivity to dexamethasone. Glutamic Acid 228-237 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 7572198-5 1995 Thus, c-fos is induced in rat striatum following administration of caffeine and other xanthines that (provided they enter the brain) block adenosine receptors, suggesting an involvement of central adenosine receptors. Xanthines 86-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 6-11 7566487-8 1995 These results show that a time-dependent and co-ordinated induction of c-fos, c-jun, jun-B and zif-268 mRNA in cerebral cortex occurs in response to the persistent excitation caused by excitotoxin injection which is mediated by glutamate and shows a differential sensitivity to dexamethasone. Dexamethasone 278-291 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 7637862-4 1995 Cannula implantation with or without 4 h of Ringer perfusion caused hardly any detectable c-fos expression in the brain, but 20 min of bicuculline (0.1 mM) perfusion induced high levels of c-fos messenger RNA and Fos protein expression in the area adjacent to the dialysis membrane, indicating activated thalamic neurons. Bicuculline 135-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 189-194 7612155-4 1995 In addition, c-fos induction in response to acute restraint stress was down-regulated by chronic, but not acute, administration of tranylcypromine or imipramine, two drugs that nonselectively increase synaptic levels of norepinephrine and serotonin by inhibition of monoamine oxidase or neurotransmitter reuptake, respectively. Norepinephrine 220-234 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 7612155-4 1995 In addition, c-fos induction in response to acute restraint stress was down-regulated by chronic, but not acute, administration of tranylcypromine or imipramine, two drugs that nonselectively increase synaptic levels of norepinephrine and serotonin by inhibition of monoamine oxidase or neurotransmitter reuptake, respectively. Serotonin 239-248 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 7637862-4 1995 Cannula implantation with or without 4 h of Ringer perfusion caused hardly any detectable c-fos expression in the brain, but 20 min of bicuculline (0.1 mM) perfusion induced high levels of c-fos messenger RNA and Fos protein expression in the area adjacent to the dialysis membrane, indicating activated thalamic neurons. Bicuculline 135-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-216 7612155-5 1995 Moreover, chronic administration of desipramine or sertraline, selective re-uptake inhibitors of norepinephrine, or serotonin, respectively, also significantly down-regulated the induction of c-fos mRNA in response to restraint stress. Desipramine 36-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 192-197 7656159-7 1995 Immunocytochemical investigation also confirmed that heparin could inhibit the expression of nuclear oncogene c-fos and c-jun in the MsC stimulated by PMA. Heparin 53-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 7612155-5 1995 Moreover, chronic administration of desipramine or sertraline, selective re-uptake inhibitors of norepinephrine, or serotonin, respectively, also significantly down-regulated the induction of c-fos mRNA in response to restraint stress. Sertraline 51-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 192-197 7612155-5 1995 Moreover, chronic administration of desipramine or sertraline, selective re-uptake inhibitors of norepinephrine, or serotonin, respectively, also significantly down-regulated the induction of c-fos mRNA in response to restraint stress. Norepinephrine 97-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 192-197 7612155-5 1995 Moreover, chronic administration of desipramine or sertraline, selective re-uptake inhibitors of norepinephrine, or serotonin, respectively, also significantly down-regulated the induction of c-fos mRNA in response to restraint stress. Serotonin 116-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 192-197 7624831-0 1995 NMDA antagonists and clonidine block c-fos expression during morphine withdrawal. N-Methylaspartate 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 7624831-0 1995 NMDA antagonists and clonidine block c-fos expression during morphine withdrawal. Clonidine 21-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 7624831-0 1995 NMDA antagonists and clonidine block c-fos expression during morphine withdrawal. Morphine 61-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 7624831-2 1995 Induction of c-fos in certain CNS regions occurs following naltrexone precipitated withdrawal in morphine dependent rats. Naltrexone 59-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 7624831-2 1995 Induction of c-fos in certain CNS regions occurs following naltrexone precipitated withdrawal in morphine dependent rats. Morphine 97-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 7624831-4 1995 We determined the levels of c-fos mRNA by solution hybridization in several brain regions in control and morphine dependent rats following pretreatment with saline, MK801 (1 mg/kg, s.c.), LY274614 (100 mg/kg, i.p. Morphine 105-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 7624831-6 1995 Morphine treatment increased c-fos mRNA levels in striatum (STR) and amygdala (AMY). Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 7624831-8 1995 However, naltrexone increased c-fos mRNA levels in morphine dependent rats in the nucleus accumbens (NA), frontal cortex (FC), AMY, and hippocampus (HIP) but not in STR or spinal cord. Naltrexone 9-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 7624831-8 1995 However, naltrexone increased c-fos mRNA levels in morphine dependent rats in the nucleus accumbens (NA), frontal cortex (FC), AMY, and hippocampus (HIP) but not in STR or spinal cord. Morphine 51-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 7656159-7 1995 Immunocytochemical investigation also confirmed that heparin could inhibit the expression of nuclear oncogene c-fos and c-jun in the MsC stimulated by PMA. Tetradecanoylphorbol Acetate 151-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 7624039-1 1995 In rats which were injected with horseradish peroxidase (HRP) into the midbrain periaqueductal gray (PAG) and then administered with formaldehyde into the stomach, Fos-like immunoreactivity was found in tyrosine hydroxylase-like immunoreactive neurons in the nucleus tractus solitarii (NTS) which were retrogradely labeled with HRP. Formaldehyde 133-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-167 7620882-2 1995 The distribution of c-fos-positive cells was dense in the superficial laminae and sparse in the deep laminae of the MDH and C1 in both young and aged animals following subcutaneous injection of formalin into the lateral face, whereas few c-fos-positive cells were labeled after saline injection. Formaldehyde 194-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 7620882-2 1995 The distribution of c-fos-positive cells was dense in the superficial laminae and sparse in the deep laminae of the MDH and C1 in both young and aged animals following subcutaneous injection of formalin into the lateral face, whereas few c-fos-positive cells were labeled after saline injection. Sodium Chloride 278-284 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 7600678-2 1995 Both c-fos and jun-B mRNA levels increased within 20 min, peaked at 40 min and declined to baseline by 120 min after cocaine treatment (30 mg/kg). Cocaine 117-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 5-10 7542145-20 1995 Further, a very large proportion of RVLM neurons that expressed c-Fos during morphine withdrawal (83%) were immunoreactive for alpha 2A-adrenergic receptors. Morphine 77-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 7614003-0 1995 Morphine, 5-HT2 and 5-HT3 receptor antagonists reduce c-fos expression in the trigeminal nuclear complex following noxious chemical stimulation of the rat nasal mucosa. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 7614003-7 1995 The reduction of the expression of Fos-like immunoreactivity by exogenous morphine speaks in favour of an opioidergic link in the modulation of orofacial pain in the trigeminal nuclei. Morphine 74-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 7614014-0 1995 Region-related Fos expression in hypothalamic dopaminergic neurons of rats by intraperitoneal administration of an endogenous satiety substance, 2-buten-4-olide. butenolide 145-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-18 7614014-1 1995 An effect of 2-buten-4-olide (2-B4O), an endogenous satiety substance, on hypothalamic dopamine neurons of rats was studied by double-label immunohistochemistry for c-Fos and tyrosine hydroxylase (TH). butenolide 13-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 7614014-1 1995 An effect of 2-buten-4-olide (2-B4O), an endogenous satiety substance, on hypothalamic dopamine neurons of rats was studied by double-label immunohistochemistry for c-Fos and tyrosine hydroxylase (TH). butenolide 30-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 7614014-2 1995 Intraperitoneal administration of 2-B4O induced Fos-like immunoreactivity in TH-immunoreactive neurons in the periventricular area and paraventricular nucleus but not in the arcuate nucleus. butenolide 34-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 7619322-4 1995 Naloxone abolished hypoalgesic responses and reinstated spinal Fos expression, indicating that aversive CSs activated opioid-based antinociceptive mechanisms. Naloxone 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 7609608-1 1995 Several studies have demonstrated that cocaine increases preprodynorphin, c-fos, and zif/268 mRNAs in rat dorsal striatum. Cocaine 39-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 7583020-2 1995 The indirect dopamine agonists, amphetamine and cocaine have been shown to induce expression of immediate early genes, such as c-fos, and neuropeptide genes, such as prodynorphin in the rat striatum. Dopamine 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 7583020-2 1995 The indirect dopamine agonists, amphetamine and cocaine have been shown to induce expression of immediate early genes, such as c-fos, and neuropeptide genes, such as prodynorphin in the rat striatum. Amphetamine 32-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 7583020-2 1995 The indirect dopamine agonists, amphetamine and cocaine have been shown to induce expression of immediate early genes, such as c-fos, and neuropeptide genes, such as prodynorphin in the rat striatum. Cocaine 48-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 7583020-4 1995 CREB interacts with functional regulatory elements in both the c-fos and prodynorphin genes, and is phosphorylated in response to dopamine in a D1 dopamine receptor-dependent manner. Dopamine 130-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 7583020-5 1995 In addition, we show by intra-striatal injection of antisense oligonucleotides directed against CREB mRNA, that CREB protein is required for c-fos induction by amphetamine. Oligonucleotides 62-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 7583020-5 1995 In addition, we show by intra-striatal injection of antisense oligonucleotides directed against CREB mRNA, that CREB protein is required for c-fos induction by amphetamine. Amphetamine 160-171 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 7789465-2 1995 Single intraperitoneal injection of carbamazepine (50 mg/kg), valproate (300 mg/kg) or intravenous injection of phenytoine (20 mg/kg) before noxious stimulation reduced the number of c-Fos immunoreactive neurons to 65-80% of control levels in superficial laminae and to 30-60% in deep laminae of the dorsal horn. Carbamazepine 36-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 183-188 7789465-2 1995 Single intraperitoneal injection of carbamazepine (50 mg/kg), valproate (300 mg/kg) or intravenous injection of phenytoine (20 mg/kg) before noxious stimulation reduced the number of c-Fos immunoreactive neurons to 65-80% of control levels in superficial laminae and to 30-60% in deep laminae of the dorsal horn. Valproic Acid 62-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 183-188 7789465-2 1995 Single intraperitoneal injection of carbamazepine (50 mg/kg), valproate (300 mg/kg) or intravenous injection of phenytoine (20 mg/kg) before noxious stimulation reduced the number of c-Fos immunoreactive neurons to 65-80% of control levels in superficial laminae and to 30-60% in deep laminae of the dorsal horn. Phenytoin 112-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 183-188 7789465-3 1995 Pretreatment with carbamazepine or valproate for 4 or 8 days combined with an injection immediately before noxious stimulation further significantly decreased the number of c-Fos neurons in the deep dorsal horn only in animals treated with valproate. Carbamazepine 18-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 173-178 7789465-3 1995 Pretreatment with carbamazepine or valproate for 4 or 8 days combined with an injection immediately before noxious stimulation further significantly decreased the number of c-Fos neurons in the deep dorsal horn only in animals treated with valproate. Valproic Acid 35-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 173-178 7789465-3 1995 Pretreatment with carbamazepine or valproate for 4 or 8 days combined with an injection immediately before noxious stimulation further significantly decreased the number of c-Fos neurons in the deep dorsal horn only in animals treated with valproate. Valproic Acid 240-249 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 173-178 7714797-6 1995 In contrast, the linkage of cAMP to cerebellar c-fos expression showed marked deficiencies in lesioned animals and a corresponding loss of the ability of beta receptors to induce c-fos; accordingly, this is a likely point at which beta adrenergic control of ODC is programmed by neuronal input. Cyclic AMP 28-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 7542145-14 1995 C-Fos expression induced by naltrexone-precipitated withdrawal was examined in the brainstem of freely moving morphine-dependent rats pretreated with clonidine or saline before injection of the opioid antagonist. Naltrexone 28-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 7542145-14 1995 C-Fos expression induced by naltrexone-precipitated withdrawal was examined in the brainstem of freely moving morphine-dependent rats pretreated with clonidine or saline before injection of the opioid antagonist. Morphine 110-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 7542145-14 1995 C-Fos expression induced by naltrexone-precipitated withdrawal was examined in the brainstem of freely moving morphine-dependent rats pretreated with clonidine or saline before injection of the opioid antagonist. Clonidine 150-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 7542145-14 1995 C-Fos expression induced by naltrexone-precipitated withdrawal was examined in the brainstem of freely moving morphine-dependent rats pretreated with clonidine or saline before injection of the opioid antagonist. Sodium Chloride 163-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 7542145-16 1995 Morphine withdrawal without clonidine treatment significantly increased the number of Fos-like-immunoreactive (Fos-LIR) cells in the RVLM and LC. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 7542145-16 1995 Morphine withdrawal without clonidine treatment significantly increased the number of Fos-like-immunoreactive (Fos-LIR) cells in the RVLM and LC. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 7705776-11 1995 In addition, under the same conditions, the EGF-induced c-fos mRNA accumulation was blocked by Dex whereas TGF-beta had no effect. Dexamethasone 95-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 7714797-8 1995 In the forebrain, for example, neonatal lesions produced receptor upregulation and supersensitivity of c-fos to cAMP stimulation. Cyclic AMP 112-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 7609873-0 1995 Receptor mechanisms mediating clozapine-induced c-fos expression in the forebrain. Clozapine 30-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 7609873-1 1995 The atypical antipsychotic clozapine produces distinctly different regional patterns of c-fos expression in rat forebrain than does the prototypical neuroleptic haloperidol. Clozapine 27-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 7609873-2 1995 While haloperidol-induced c-fos expression appears to be mediated by its D2 dopamine receptor antagonist properties, the mechanisms by which clozapine increases c-fos expression remain uncertain. Haloperidol 6-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 7609873-2 1995 While haloperidol-induced c-fos expression appears to be mediated by its D2 dopamine receptor antagonist properties, the mechanisms by which clozapine increases c-fos expression remain uncertain. Clozapine 141-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 7609873-3 1995 Using a combination of brain lesion, pharmacological and immunohistochemical techniques, the present study sought to determine the receptor mechanisms by which clozapine increases the number of Fos-like immunoreactive neurons in various regions of the forebrain. Clozapine 160-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 194-197 7609873-4 1995 To test whether serotonergic and/or noradrenergic systems are involved in clozapine-induced c-fos expression, rats received either 5,7-dihydroxytryptamine lesions of the medial forebrain bundle or 6-hydroxydopamine lesions of the dorsal noradrenergic bundle two weeks prior to clozapine (20 mg/kg) injections. Clozapine 74-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 7609873-9 1995 Quinpirole produced a small but significant decrease in clozapine-induced c-fos expression in the medial prefrontal cortex, had larger effects in the lateral septum, and blocked clozapine"s actions in the nucleus accumbens and major island of Calleja. Quinpirole 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 7609873-9 1995 Quinpirole produced a small but significant decrease in clozapine-induced c-fos expression in the medial prefrontal cortex, had larger effects in the lateral septum, and blocked clozapine"s actions in the nucleus accumbens and major island of Calleja. Clozapine 56-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 7609873-10 1995 Pretreatment with 7-OH-DPAT attenuated clozapine-induced c-fos expression in the nucleus accumbens and lateral septum, completely blocked the expression in the major island of Calleja, but was without effect in the medial prefrontal cortex. 7-hydroxy-2-N,N-dipropylaminotetralin 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 7609873-10 1995 Pretreatment with 7-OH-DPAT attenuated clozapine-induced c-fos expression in the nucleus accumbens and lateral septum, completely blocked the expression in the major island of Calleja, but was without effect in the medial prefrontal cortex. Clozapine 39-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 7609873-11 1995 Given the different affinities of quinpirole and 7-OH-DPAT for D2, D3 and D4 receptors, these data suggest that clozapine-induced increases in c-fos expression in the nucleus accumbens, major island of Cajella and lateral septal nucleus are due to antagonist actions of this antipsychotic at D3 dopamine receptors. Quinpirole 34-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-148 7609873-11 1995 Given the different affinities of quinpirole and 7-OH-DPAT for D2, D3 and D4 receptors, these data suggest that clozapine-induced increases in c-fos expression in the nucleus accumbens, major island of Cajella and lateral septal nucleus are due to antagonist actions of this antipsychotic at D3 dopamine receptors. 7-hydroxy-2-N,N-dipropylaminotetralin 49-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-148 7609873-11 1995 Given the different affinities of quinpirole and 7-OH-DPAT for D2, D3 and D4 receptors, these data suggest that clozapine-induced increases in c-fos expression in the nucleus accumbens, major island of Cajella and lateral septal nucleus are due to antagonist actions of this antipsychotic at D3 dopamine receptors. Clozapine 112-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-148 7609873-12 1995 They also indicate that while antagonist actions at D4 receptors may contribute, the primary mechanisms by which clozapine increases c-fos expression in the medial prefrontal cortex remain to be determined. Clozapine 113-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 7617159-1 1995 The role of vesicular and newly synthesized dopamine in the action of amphetamine was investigated by studying the effect of reserpine and alpha-methyl-p-tyrosine pretreatment on amphetamine-induced changes in extracellular dopamine and acetylcholine, estimated by brain microdialysis, and on c-fos expression, estimated by quantitative immunohistochemistry of the Fos antigene, in the dorsal caudate-putamen of rats. Amphetamine 70-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 293-298 7617159-1 1995 The role of vesicular and newly synthesized dopamine in the action of amphetamine was investigated by studying the effect of reserpine and alpha-methyl-p-tyrosine pretreatment on amphetamine-induced changes in extracellular dopamine and acetylcholine, estimated by brain microdialysis, and on c-fos expression, estimated by quantitative immunohistochemistry of the Fos antigene, in the dorsal caudate-putamen of rats. Amphetamine 70-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 365-368 7617159-5 1995 alpha-Methyl-p-tyrosine pretreatment reduced c-fos expression stimulated by amphetamine (2 mg/kg) in the dorsomedial and dorsolateral caudate-putamen while reserpine pretreatment reduced it only in the dorsolateral caudate-putamen. alpha-Methyltyrosine 0-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 7617159-5 1995 alpha-Methyl-p-tyrosine pretreatment reduced c-fos expression stimulated by amphetamine (2 mg/kg) in the dorsomedial and dorsolateral caudate-putamen while reserpine pretreatment reduced it only in the dorsolateral caudate-putamen. Amphetamine 76-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 7617160-0 1995 Acute and chronic amphetamine treatments differently regulate neuropeptide messenger RNA levels and Fos immunoreactivity in rat striatal neurons. Amphetamine 18-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-103 7617160-7 1995 A double labeling procedure with Fos immunohistochemistry coupled with in situ hybridization demonstrated that acute amphetamine treatment induces Fos immunoreactivity predominantly in striatal neurons expressing substance P messenger RNA (77.07 +/- 1.42%). Amphetamine 117-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 7617160-7 1995 A double labeling procedure with Fos immunohistochemistry coupled with in situ hybridization demonstrated that acute amphetamine treatment induces Fos immunoreactivity predominantly in striatal neurons expressing substance P messenger RNA (77.07 +/- 1.42%). Amphetamine 117-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-150 7617160-9 1995 In chronic amphetamine treated rats, 56.21 +/- 1.32% of the Fos immunoreactive neurons expressed substance P messenger RNA while 52.12 +/- 1.84% expressed preproenkephalin A messenger RNA. Amphetamine 11-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 7617160-11 1995 Amphetamine treatments induced Fos immunoreactivity in the substantia nigra in non-dopamine neurons. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 7718243-5 1995 Surprisingly, following chronic administration of amphetamine, levels of phosphorylated CREB are increased above basal in rat striatum in vivo, whereas c-fos mRNA is suppressed below basal levels. Amphetamine 50-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-157 7783858-8 1995 In situ hybridization was performed by hybridizing sections with 35S-labeled c-fos cRNA probes. Sulfur-35 65-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 7783858-9 1995 ICV injection of vehicle alone induced a weak c-fos mRNA expression in the lateral septal nucleus (LSV) and PVN in the rats without restraint, probably due to the mild stress of ICV injection. icv 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 7566476-0 1995 Induction of c-fos, jun B and egr-1 expression by haloperidol in PC12 cells: involvement of calcium. Haloperidol 50-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 7783858-9 1995 ICV injection of vehicle alone induced a weak c-fos mRNA expression in the lateral septal nucleus (LSV) and PVN in the rats without restraint, probably due to the mild stress of ICV injection. icv 178-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 7566476-1 1995 Acute injection of haloperidol, a dopamine D2 receptor antagonist, is known to increase immediate early gene expression of the fos and jun families in rodent striatal neurons. Haloperidol 19-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-130 7566476-2 1995 A set of gene induction, including c-fos, jun B and TIS8/egr-1, was found when haloperidol was added to PC12 cells in culture. Haloperidol 79-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 7792718-2 1995 Local infusion into the SN of the 6-OHDA lesioned side of MK 801, CPP or NBQX at doses inducing no or minimal behavioral effects significantly increased the turning behavior and the expression of c-fos induced, in the lesioned caudate-putamen (CPu), by a parenteral administration of SKF 38393. Oxidopamine 34-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 196-201 7792718-2 1995 Local infusion into the SN of the 6-OHDA lesioned side of MK 801, CPP or NBQX at doses inducing no or minimal behavioral effects significantly increased the turning behavior and the expression of c-fos induced, in the lesioned caudate-putamen (CPu), by a parenteral administration of SKF 38393. Dizocilpine Maleate 58-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 196-201 7792718-2 1995 Local infusion into the SN of the 6-OHDA lesioned side of MK 801, CPP or NBQX at doses inducing no or minimal behavioral effects significantly increased the turning behavior and the expression of c-fos induced, in the lesioned caudate-putamen (CPu), by a parenteral administration of SKF 38393. 2,3-dioxo-6-nitro-7-sulfamoylbenzo(f)quinoxaline 73-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 196-201 7792718-2 1995 Local infusion into the SN of the 6-OHDA lesioned side of MK 801, CPP or NBQX at doses inducing no or minimal behavioral effects significantly increased the turning behavior and the expression of c-fos induced, in the lesioned caudate-putamen (CPu), by a parenteral administration of SKF 38393. 1,2,3,4-tetrahydroisoquinoline-7-sulfonamide 284-287 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 196-201 7792718-4 1995 High doses of MK 801, CPP or muscimol infused into the SN produced intense contralateral turning per se and induced a sparse c-fos expression in the lesioned CPu which was antagonized by parenteral administration of MK 801. Dizocilpine Maleate 14-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 7792718-4 1995 High doses of MK 801, CPP or muscimol infused into the SN produced intense contralateral turning per se and induced a sparse c-fos expression in the lesioned CPu which was antagonized by parenteral administration of MK 801. Muscimol 29-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 7792718-4 1995 High doses of MK 801, CPP or muscimol infused into the SN produced intense contralateral turning per se and induced a sparse c-fos expression in the lesioned CPu which was antagonized by parenteral administration of MK 801. Dizocilpine Maleate 216-222 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 7605952-3 1995 In this study, antisense oligonucleotides complementary to c-fos mRNA were employed to interfere with the expression of Fos protein. Oligonucleotides 25-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 7605952-3 1995 In this study, antisense oligonucleotides complementary to c-fos mRNA were employed to interfere with the expression of Fos protein. Oligonucleotides 25-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-123 7605952-4 1995 Injections in the rat medial preoptic area of c-fos antisense, but not of sense, oligonucleotides blocked the expression of Fos protein detected immunocytochemically. Oligonucleotides 81-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 7796106-1 1995 c-fos immunoreactivity was used to map brain areas in which neurons reacted either to electrical stimulation or to microinjection of the excitatory amino acid kainate and of the GABAA antagonist, SR-95531, applied to the medial hypothalamus of freely moving rats. excitatory amino acid kainate 137-166 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 7609902-0 1995 Concurrent reduction of inflammation and spinal Fos-LI neurons by systemic diclofenac in the rat. Diclofenac 75-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 7609902-1 1995 Intraplantar carrageenan induced a peripheral inflammation and c-Fos like immunoreactivity (Fos-LI) in the dorsal horn, L4-L5 segments, of the rat spinal cord. Carrageenan 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 7609902-1 1995 Intraplantar carrageenan induced a peripheral inflammation and c-Fos like immunoreactivity (Fos-LI) in the dorsal horn, L4-L5 segments, of the rat spinal cord. Carrageenan 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 7609902-2 1995 The total number of control carrageenan evoked Fos-LI neurons was 126 +/- 6 per section, which were predominately located in the superficial and deep laminae (41 +/- 2% and 39 +/- 2% of the total number of Fos-LI neurons per section) of the dorsal horn. Carrageenan 28-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 7890729-5 1995 Nuclear run-on assays indicated that okadaic acid also activated NGF gene transcription, which was preceded by an induction of c-fos and c-jun gene transcription. Okadaic Acid 37-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 7796106-1 1995 c-fos immunoreactivity was used to map brain areas in which neurons reacted either to electrical stimulation or to microinjection of the excitatory amino acid kainate and of the GABAA antagonist, SR-95531, applied to the medial hypothalamus of freely moving rats. gabazine 196-204 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 7624023-0 1995 A subset of striatopallidal neurons are Fos-immunopositive following acute monoamine depletion in the rat. monoamine 75-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 7773694-1 1995 Experiments were done in conscious rats to investigate the effect of intravenous infusion of hypertonic saline on the induction of the phosphoprotein Fos in brainstem catecholaminergic neurons. Sodium Chloride 104-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-153 7773694-3 1995 Infusions of 165 mM or 1.4 M NaCl solutions into the jugular vein resulted in Fos-like immunoreactivity in approximately the caudal two thirds of nucleus of the solitary tract (NTS), the caudal and rostral ventrolateral medulla (VLM), and in the lateral aspects of the parabrachial nucleus (PBN). Sodium Chloride 29-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 7624023-1 1995 Experiments were conducted to characterise the Fos-immunopositive neurons that are observed in the dorsal rim of the striatum following monoamine depletion by the systemic administration of reserpine. monoamine 136-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 7624023-1 1995 Experiments were conducted to characterise the Fos-immunopositive neurons that are observed in the dorsal rim of the striatum following monoamine depletion by the systemic administration of reserpine. Reserpine 190-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 7624023-4 1995 It can be concluded that Fos levels are increased only in a subset of striatopallidal neurons following monoamine depletion. monoamine 104-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 7745131-4 1995 In a prior study, we have shown that both regional and temporal patterns of c-fos induction following treatment with the indirect dopamine receptor agonist cocaine are inversely related to those of dynorphin expression. Cocaine 156-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 7890661-6 1995 The down-regulation of gax in response to balloon injury mirrors the up-regulation seen in a number of early response genes such as c-myc and c-fos. 1-{3-[(4-Pyridin-2-Ylpiperazin-1-Yl)sulfonyl]phenyl}-3-(1,3-Thiazol-2-Yl)urea 23-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-147 7477877-6 1995 It was demonstrated that in drug-naive rats bearing unilateral 6-hydroxydopamine lesions of the dopaminergic nigrostriatal pathway, the mixed D1/D2 agonist apomorphine produced a dramatic increase in the expression of Fos-like immunoreactivity in the ipsilateral caudoputamen, nucleus accumbens and globus pallidus, and was a potent primer of SKF-38393-mediated rotational behaviour. Oxidopamine 63-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 218-221 7477877-6 1995 It was demonstrated that in drug-naive rats bearing unilateral 6-hydroxydopamine lesions of the dopaminergic nigrostriatal pathway, the mixed D1/D2 agonist apomorphine produced a dramatic increase in the expression of Fos-like immunoreactivity in the ipsilateral caudoputamen, nucleus accumbens and globus pallidus, and was a potent primer of SKF-38393-mediated rotational behaviour. Apomorphine 156-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 218-221 7477877-8 1995 Preadministration of MK-801 at 0.5 mg/kg significantly reduced apomorphine"s effect on Fos expression and prevented apomorphine priming of SKF-38393-induced rotation. Dizocilpine Maleate 21-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 7477877-8 1995 Preadministration of MK-801 at 0.5 mg/kg significantly reduced apomorphine"s effect on Fos expression and prevented apomorphine priming of SKF-38393-induced rotation. Apomorphine 63-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 7477877-10 1995 In another experiment, the D2 family-selective agonist quinpirole was found to be an affective primer of SKF-38393-mediated rotation, and to produce increase Fos expression in the ipsilateral globus pallidus only. Quinpirole 55-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-161 7477877-11 1995 Preadministration of MK-801 at 0.1 mg/kg blocked quinpirole priming of SKF-38393-mediated rotation and significantly reduced the number of Fos-positive neurons in the ipsilateral globus pallidus. Dizocilpine Maleate 21-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 7477877-12 1995 Administration of the indirect dopamine agonist amphetamine increased Fos expression in the intact striatum, but not in the ipsilateral (lesioned) striatum or globus pallidus, and did not sensitize (prime) animals to behavioural effects of SKF-38393. Dopamine 31-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 7477877-12 1995 Administration of the indirect dopamine agonist amphetamine increased Fos expression in the intact striatum, but not in the ipsilateral (lesioned) striatum or globus pallidus, and did not sensitize (prime) animals to behavioural effects of SKF-38393. Amphetamine 48-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 7477877-14 1995 Northern blot analysis demonstrated that a priming dose of apomorphine significantly increased the messenger RNA signals for c-fos, c-jun, ngfi-A and jun-B in denervated striatum. Apomorphine 59-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 7477881-6 1995 In further experiments, rhodamine-conjugated latex microspheres were injected into the supraoptic nucleus to retrogradely label afferent neurons, and the brains were processed with double-immunohistochemistry for tyrosine hydroxylase and Fos-like protein. Rhodamines 24-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 238-241 7777163-0 1995 c-fos antisense oligodeoxynucleotide increases formalin-induced nociception and regulates preprodynorphin expression. Oligodeoxyribonucleotides 16-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 7900910-1 1995 Systemic injection of the fructose analogue 2,5-anhydro-D-mannitol (2,5-AM) elicits a feeding response and induces c-fos activity in the parabrachial nuclei (PBN). Fructose 26-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 7900910-1 1995 Systemic injection of the fructose analogue 2,5-anhydro-D-mannitol (2,5-AM) elicits a feeding response and induces c-fos activity in the parabrachial nuclei (PBN). 2,5-anhydromannitol 44-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 7900910-1 1995 Systemic injection of the fructose analogue 2,5-anhydro-D-mannitol (2,5-AM) elicits a feeding response and induces c-fos activity in the parabrachial nuclei (PBN). 2,5-anhydromannitol 68-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 7769993-0 1995 Time course of phosphorylated CREB and Fos-like immunoreactivity in the hypothalamic supraoptic nucleus after salt loading. Salts 110-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 7769993-4 1995 Forty-five minutes after injection, levels of c-fos mRNA in the SON were elevated in hypertonic saline-treated rats as compared with normal saline-treated rats, and were minimally detectable in uninjected rats. Sodium Chloride 96-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 7769993-5 1995 At this time period, the hypertonic saline-treated rats showed increased number of Fos-LI cells in the SON, whereas normal saline-treated rats showed little or no Fos-LI cells. hypertonic 25-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 7769993-5 1995 At this time period, the hypertonic saline-treated rats showed increased number of Fos-LI cells in the SON, whereas normal saline-treated rats showed little or no Fos-LI cells. Sodium Chloride 36-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 7769993-5 1995 At this time period, the hypertonic saline-treated rats showed increased number of Fos-LI cells in the SON, whereas normal saline-treated rats showed little or no Fos-LI cells. Sodium Chloride 36-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 7769993-6 1995 The discrepancy between levels of PCREB-LI and c-fos mRNA suggests that injection of hypertonic saline may activate additional transcriptional factors besides CREB. Sodium Chloride 96-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 7605896-13 1995 In the isolated perfused working rat heart, norepinephrine (3 x 10(-8) M) increased the expression of the proto-oncogenes c-fos and c-myc after 30 and 60 minutes, respectively. Norepinephrine 44-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 7773436-6 1995 In contrast, light pulses with or without the control nonsense oligodeoxynucleotides evoked strong nuclear c-Fos and JunB immunoreactivity in the rat suprachiasmatic nucleus. Oligodeoxyribonucleotides 63-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 7891165-9 1995 Thus, treatment with behaviorally effective doses of both estradiol and progesterone induces Fos expression in localized regions of female rat brain. Estradiol 58-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 7891165-9 1995 Thus, treatment with behaviorally effective doses of both estradiol and progesterone induces Fos expression in localized regions of female rat brain. Progesterone 72-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 7891173-6 1995 cAMP analogs and forskolin induced widespread expression of both Fos-like and Fra-like proteins. Cyclic AMP 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 7891173-6 1995 cAMP analogs and forskolin induced widespread expression of both Fos-like and Fra-like proteins. Colforsin 17-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 7596621-4 1995 The expression of the Fos protein was related to the intensity of stimulation since greater injection volumes of formalin or prolonged application of the thermal stimulus increased the number of stained nuclei. Formaldehyde 113-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-25 7540319-6 1995 In contrast, mRNA levels for c-fos were dramatically elevated in the LC following naloxone-precipitated withdrawal. Naloxone 82-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 7864828-0 1995 Analysis of c-fos expression in the butyrate-induced F-98 glioma cell differentiation. Butyrates 36-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 7864828-1 1995 The functional induction of c-fos in the sodium butyrate-induced differentiation of F-98 glioma cells was studied. Butyric Acid 41-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 7864828-2 1995 Fos protein level was increased by butyrate. Butyrates 35-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 7864828-4 1995 Gel-retardation assay indicates Fos as a component of the complex formed between the consensus oligonucleotide of the TPA (PMA, phorbol 12-myristate 13-acetate) response element (TRE) and nuclear extract prepared from butyrate-treated cells. Oligonucleotides 95-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 7864828-4 1995 Gel-retardation assay indicates Fos as a component of the complex formed between the consensus oligonucleotide of the TPA (PMA, phorbol 12-myristate 13-acetate) response element (TRE) and nuclear extract prepared from butyrate-treated cells. Tetradecanoylphorbol Acetate 118-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 7864828-4 1995 Gel-retardation assay indicates Fos as a component of the complex formed between the consensus oligonucleotide of the TPA (PMA, phorbol 12-myristate 13-acetate) response element (TRE) and nuclear extract prepared from butyrate-treated cells. Tetradecanoylphorbol Acetate 123-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 7864828-4 1995 Gel-retardation assay indicates Fos as a component of the complex formed between the consensus oligonucleotide of the TPA (PMA, phorbol 12-myristate 13-acetate) response element (TRE) and nuclear extract prepared from butyrate-treated cells. Tetradecanoylphorbol Acetate 128-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 7864828-4 1995 Gel-retardation assay indicates Fos as a component of the complex formed between the consensus oligonucleotide of the TPA (PMA, phorbol 12-myristate 13-acetate) response element (TRE) and nuclear extract prepared from butyrate-treated cells. Butyrates 218-226 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 7864828-5 1995 Transfection studies showed that butyrate increased transcription from a multimeric TRE-driven reporter construct, and the effect was mimicked by transfecting cells with fos-expression plasmid. Butyrates 33-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-173 7864828-6 1995 Furthermore, under conditions of c-fos over-expression, transactivation by butyrate was essentially abolished. Butyrates 75-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 7864828-10 1995 Transfection of the c-fos promoter-chloramphenicol acetyltransferase fusion gene into F-98 cells revealed that activation of c-fos by butyrate was exerted at the promoter level, and sequences located within nucleotides -757 to -402 of the c-fos promoter were responsible for butyrate induction. Butyrates 134-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 7864828-10 1995 Transfection of the c-fos promoter-chloramphenicol acetyltransferase fusion gene into F-98 cells revealed that activation of c-fos by butyrate was exerted at the promoter level, and sequences located within nucleotides -757 to -402 of the c-fos promoter were responsible for butyrate induction. Butyrates 134-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 7864828-10 1995 Transfection of the c-fos promoter-chloramphenicol acetyltransferase fusion gene into F-98 cells revealed that activation of c-fos by butyrate was exerted at the promoter level, and sequences located within nucleotides -757 to -402 of the c-fos promoter were responsible for butyrate induction. Butyrates 134-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 7864828-10 1995 Transfection of the c-fos promoter-chloramphenicol acetyltransferase fusion gene into F-98 cells revealed that activation of c-fos by butyrate was exerted at the promoter level, and sequences located within nucleotides -757 to -402 of the c-fos promoter were responsible for butyrate induction. Butyrates 275-283 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 7864828-10 1995 Transfection of the c-fos promoter-chloramphenicol acetyltransferase fusion gene into F-98 cells revealed that activation of c-fos by butyrate was exerted at the promoter level, and sequences located within nucleotides -757 to -402 of the c-fos promoter were responsible for butyrate induction. Butyrates 275-283 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 7864828-10 1995 Transfection of the c-fos promoter-chloramphenicol acetyltransferase fusion gene into F-98 cells revealed that activation of c-fos by butyrate was exerted at the promoter level, and sequences located within nucleotides -757 to -402 of the c-fos promoter were responsible for butyrate induction. Butyrates 275-283 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 7864828-11 1995 Our data indicate that transcriptional activation of c-fos through its promoter by butyrate resulted in increased Fos protein expression. Butyrates 83-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 7753494-7 1995 Distribution of FOS-ir neurons in the DMX corresponds with that of the visceromotor neurons involved in gastric secretion. dmx 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 7776969-6 1995 The mitogenic effect of TGF-beta was abolished in cells cultured in the presence of AS, whereas S had no effect, showing that c-fos is required for TGF beta-induced osteoblast cell growth. Arsenic 84-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 7784901-0 1995 [Morphine-3-glucuronide (M3G) potentiates noxious stimulus-evoked Fos protein-like immunoreactivity in the rat spinal cord]. morphine-3-glucuronide 1-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 7784901-1 1995 The effect of morphine-3-glucuronide (M3G) on noxious stimulus-evoked Fos protein-like immunoreactivity in the rat spinal cord were assessed by ABC method. morphine-3-glucuronide 14-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 7784901-2 1995 It was found that a dose-dependent increase of Fos-like immunoreactive neurons could be induced by M3G intrathecal injection followed by formaline injection into hindpaw. Formaldehyde 137-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 7540119-3 1995 It was also demonstrated that tumor promoter phenobarbital influenced c-fos and c-myc expressions and decreased alpha 1I3 mRNA level in rat liver during a long term experiment. Phenobarbital 45-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 7743187-0 1995 Dopamine antagonists induce fos-like-immunoreactivity in the substantia nigra and entopeduncular nucleus of the rat. Dopamine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 7743187-1 1995 Injections of the D2 receptor antagonists haloperidol (0.5-8 mg/kg) and metoclopramide (6.25-50 mg/kg) in rats resulted in a dose dependent induction of Fos-like-immunoreactivity in the rostral portion of the entopeduncular nucleus (EPN) and in the medial portion of the pars reticulata of the substantia nigra (SNpr). Haloperidol 42-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-156 7743187-1 1995 Injections of the D2 receptor antagonists haloperidol (0.5-8 mg/kg) and metoclopramide (6.25-50 mg/kg) in rats resulted in a dose dependent induction of Fos-like-immunoreactivity in the rostral portion of the entopeduncular nucleus (EPN) and in the medial portion of the pars reticulata of the substantia nigra (SNpr). Metoclopramide 72-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-156 7737311-0 1995 Differential Fos-protein induction in rat forebrain regions after acute and long-term haloperidol and clozapine treatment. Haloperidol 86-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 7737311-0 1995 Differential Fos-protein induction in rat forebrain regions after acute and long-term haloperidol and clozapine treatment. Clozapine 102-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 7737311-1 1995 Both acute and long-term effects of haloperidol and clozapine on Fos-like immunoreactive nuclei in several rat forebrain areas were quantified. Haloperidol 36-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 7737311-1 1995 Both acute and long-term effects of haloperidol and clozapine on Fos-like immunoreactive nuclei in several rat forebrain areas were quantified. Clozapine 52-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 7737311-6 1995 A single dose of haloperidol produced large increases in Fos-like immunoreactive nuclei in the striatum, the nucleus accumbens and central amygdala. Haloperidol 17-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 7737311-8 1995 Acute clozapine treatment had slight (if any) effects on the number of Fos-like immunoreactivity-expressing nuclei in the striatum, but the increases in the nucleus accumbens, the lateral septum, the paraventricular and supraoptic nuclei of the hypothalamus and the central amygdala were substantial. Clozapine 6-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 7756634-2 1995 Microinjections of NPY and l-adrenaline alone into the NTS induced neuronal c-Fos IR in parts of the NTS-dmnX complex, while clonidine had no action. Epinephrine 27-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 7756634-2 1995 Microinjections of NPY and l-adrenaline alone into the NTS induced neuronal c-Fos IR in parts of the NTS-dmnX complex, while clonidine had no action. dmnx 105-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 7756634-4 1995 Coinjections of NPY and l-adrenaline or clonidine into the NTS reduced the NPY-induced increase in c-Fos IR. Epinephrine 24-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 7756634-4 1995 Coinjections of NPY and l-adrenaline or clonidine into the NTS reduced the NPY-induced increase in c-Fos IR. Clonidine 40-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 7756634-5 1995 The results emphasize that during cardiovascular regulation NPY receptors can control neuronal gene expression in the NTS-dmnX complex via c-fos and that stimulation of alpha 2-adrenoceptors can attenuate NPY receptor transduction also in terms of induction of c-Fos IR in the NTS-dmnX complex via an antagonistic alpha 2/NPY receptor interaction. dmnx 122-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 7713169-0 1995 Carrageenin-evoked c-Fos expression in rat lumbar spinal cord: the effects of indomethacin. Carrageenan 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 7713169-1 1995 This study evaluated the effects of systemic indomethacin on carrageenin evoked c-Fos expression in rat lumbar spinal cord neurons. Indomethacin 45-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 7713169-1 1995 This study evaluated the effects of systemic indomethacin on carrageenin evoked c-Fos expression in rat lumbar spinal cord neurons. Carrageenan 61-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 7713169-2 1995 Fos-like immunoreactivity was not observed after the intraplantar injection of the control vehicle saline. Sodium Chloride 99-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 7713169-3 1995 2 h after administration of carrageenin (6 mg/150 microliters) into the hind limb, Fos-like immunoreactive neurons were observed in the lumbar spinal cord (64 labelled neurons per L4-L5 sections) and were numerous in the superficial laminae (I-II), whereas at 3-4 h both superficial and deeper laminae (V, VI and ventral horn) were labelled. Carrageenan 28-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 7713169-4 1995 3 h after carrageenin administration, maximal Fos-like immunoreactivity was observed (104 labelled neurons per L4-L5 sections). Carrageenan 10-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 7713169-8 1995 With increasing doses of carrageenin, an increase in the number of Fos-like immunoreactive neurons was observed. Carrageenan 25-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 7713169-9 1995 The number of Fos-like immunoreactive neurons induced by the carrageenin stimulation (6 mg, at 3 h) was clearly reduced by oral pretreatment with indomethacin (20 mg/kg). Carrageenan 61-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 7713169-9 1995 The number of Fos-like immunoreactive neurons induced by the carrageenin stimulation (6 mg, at 3 h) was clearly reduced by oral pretreatment with indomethacin (20 mg/kg). Indomethacin 146-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 7713169-11 1995 indomethacin (1, 2.5 or 5 mg/kg) dose dependently reduced the number of Fos-like immunoreactive neurons and the inflammation of the paw and the ankle of the injected foot. Indomethacin 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 7713169-12 1995 A strong relationship between the effect of indomethacin on c-Fos expression and its effect on inflammatory processes was observed. Indomethacin 44-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 7713169-13 1995 These results suggest that Fos-like immunoreactivity induced by carrageenin inflammation may be a very useful tool to study the effects of anti-inflammatory drugs, at both peripheral and central levels of inflammation. Carrageenan 64-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 7534774-0 1995 Substance P phenotype defines specificity of c-fos induction by cocaine in developing rat striatum. Cocaine 64-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 7534774-3 1995 At each age examined, substance P+, enkephalin- striatal neurons were the predominant class of cells in which cocaine induced c-fos gene expression. Cocaine 110-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 7746492-1 1995 Staining of c-fos-like-immunoreactivity (CFIR) in neurones was used to study neuronal activation within subdivisions of periaqueductal gray (PAG), and in locus coeruleus and ventral tegmental area during opiate withdrawal in awake and anaesthetised, morphine-dependent rats. Opiate Alkaloids 204-210 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 7746492-1 1995 Staining of c-fos-like-immunoreactivity (CFIR) in neurones was used to study neuronal activation within subdivisions of periaqueductal gray (PAG), and in locus coeruleus and ventral tegmental area during opiate withdrawal in awake and anaesthetised, morphine-dependent rats. Morphine 250-258 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 7746492-6 1995 Induction of c-fos in lateral and ventrolateral PAG during withdrawal is consistent with known functions of these regions, involving the integration of autonomic and somatic components of defensive and escape behaviours which are characteristic signs of opiate withdrawal. Opiate Alkaloids 254-260 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 7896938-6 1995 We report that, during rat brain development, cocaine produced brain region-specific and developmental age-specific induction of c-fos, c-jun, and zif/268 mRNAs. Cocaine 46-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 7896938-7 1995 At each age studied (P8, P15, P28, and adults), we found that acute cocaine administration resulted in a unique cell-specific pattern of c-fos mRNA induction and c-Fos protein expression in striatum. Cocaine 68-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 7896938-7 1995 At each age studied (P8, P15, P28, and adults), we found that acute cocaine administration resulted in a unique cell-specific pattern of c-fos mRNA induction and c-Fos protein expression in striatum. Cocaine 68-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 7896938-8 1995 We also observed cocaine-induced activation of AP-1 DNA binding activity in striatal extracts prepared at these different ages, suggesting that the observed induction of c-fos and c-jun may have biological consequences for the developing brain. Cocaine 17-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-175 7732785-0 1995 Periodic acid-Schiff (PAS)-positive deposits in brain following kainic acid-induced seizures: relationships to fos induction, neuronal necrosis, reactive gliosis, and blood-brain barrier breakdown. periodic acid-schiff 0-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 7732785-0 1995 Periodic acid-Schiff (PAS)-positive deposits in brain following kainic acid-induced seizures: relationships to fos induction, neuronal necrosis, reactive gliosis, and blood-brain barrier breakdown. Aminosalicylic Acid 22-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 7668153-0 1995 Chronic treatment with morphine and ethanol, but not cocaine, attenuates IL-1 beta activation of FOS expression in the rat hypothalamic paraventricular nucleus. Morphine 23-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 7668153-0 1995 Chronic treatment with morphine and ethanol, but not cocaine, attenuates IL-1 beta activation of FOS expression in the rat hypothalamic paraventricular nucleus. Ethanol 36-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 7668153-5 1995 In this study, we compared the effects of morphine, ethanol and cocaine on IL-1 beta induction of FOS-IR in the rat hypothalamus. Morphine 42-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 7668153-5 1995 In this study, we compared the effects of morphine, ethanol and cocaine on IL-1 beta induction of FOS-IR in the rat hypothalamus. Ethanol 52-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 7668153-5 1995 In this study, we compared the effects of morphine, ethanol and cocaine on IL-1 beta induction of FOS-IR in the rat hypothalamus. Cocaine 64-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 7668153-6 1995 Acute treatment with morphine or ethanol induced FOS-IR in several nuclei including the PVN. Morphine 21-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 7668153-6 1995 Acute treatment with morphine or ethanol induced FOS-IR in several nuclei including the PVN. Ethanol 33-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 7668153-7 1995 Cocaine, which induced FOS-IR in the Caudate-Putamen (CPU), nucleus Accumbens (nAcc) and Locus Coeruleus (LC), however, did not induce FOS-IR in the PVN. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 7668153-8 1995 Chronic treatment with morphine desensitized FOS responsiveness to morphine and IL-1 beta in the PVN since FOS-IR was no longer induced by IL-1 beta or morphine in the PVN after this treatment. Morphine 23-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 7668153-8 1995 Chronic treatment with morphine desensitized FOS responsiveness to morphine and IL-1 beta in the PVN since FOS-IR was no longer induced by IL-1 beta or morphine in the PVN after this treatment. Morphine 23-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 7668153-8 1995 Chronic treatment with morphine desensitized FOS responsiveness to morphine and IL-1 beta in the PVN since FOS-IR was no longer induced by IL-1 beta or morphine in the PVN after this treatment. Morphine 67-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 7668153-8 1995 Chronic treatment with morphine desensitized FOS responsiveness to morphine and IL-1 beta in the PVN since FOS-IR was no longer induced by IL-1 beta or morphine in the PVN after this treatment. Morphine 67-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 7668153-9 1995 Similarly, chronic ethanol treatment desensitized FOS responsiveness to ethanol and to IL-1 beta in the PVN. Ethanol 19-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 7668153-9 1995 Similarly, chronic ethanol treatment desensitized FOS responsiveness to ethanol and to IL-1 beta in the PVN. Ethanol 72-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 7900855-10 1995 Finally, ANG II preincubation increased alpha 1-AR agonist phenylephrine-stimulated expression of the c-fos gene. Phenylephrine 59-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 7703210-10 1995 Furthermore, topical application of a c-fos antisense oligodeoxynucleotide to the ultraviolet exposed rat eye inhibited the increase in c-Fos expression in the cornea, suggesting therapeutic activity of antisense drugs in corneal malignant and infectious diseases. Oligodeoxyribonucleotides 54-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 7703210-10 1995 Furthermore, topical application of a c-fos antisense oligodeoxynucleotide to the ultraviolet exposed rat eye inhibited the increase in c-Fos expression in the cornea, suggesting therapeutic activity of antisense drugs in corneal malignant and infectious diseases. Oligodeoxyribonucleotides 54-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 7891139-11 1995 Pretreatment of the rats with chlorisondamine alone or in combination with atropine diminished the capsaicin-induced increase in c-fos, whereas atropine alone was less efficient. Chlorisondamine 30-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 7891139-11 1995 Pretreatment of the rats with chlorisondamine alone or in combination with atropine diminished the capsaicin-induced increase in c-fos, whereas atropine alone was less efficient. Atropine 75-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 7891139-11 1995 Pretreatment of the rats with chlorisondamine alone or in combination with atropine diminished the capsaicin-induced increase in c-fos, whereas atropine alone was less efficient. Capsaicin 99-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 7891165-0 1995 Progesterone enhances an estradiol-induced increase in Fos immunoreactivity in localized regions of female rat forebrain. Progesterone 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 7891165-0 1995 Progesterone enhances an estradiol-induced increase in Fos immunoreactivity in localized regions of female rat forebrain. Estradiol 25-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 7891165-2 1995 Although treatment with high doses of estradiol has been shown to increase immunostaining for the Fos protein, an immediate early gene product that is expressed upon cellular activation, another report conflicts with this finding. Estradiol 38-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 7891165-5 1995 In experiment one, we found that treatment with 5 micrograms of estradiol increased Fos immunoreactivity (Fos-IR) within a section of the medial preoptic area and the dorsal medial hypothalamus. Estradiol 64-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 7891165-5 1995 In experiment one, we found that treatment with 5 micrograms of estradiol increased Fos immunoreactivity (Fos-IR) within a section of the medial preoptic area and the dorsal medial hypothalamus. Estradiol 64-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 7784961-3 1995 This study illustrates how a 2 week, twice daily 7.5 mg/kg d-amphetamine or saline regimen alters rat brain regional expression of transcription factor genes, including c-fos, fos-B, jun-B, c-jun, and zif 268, and seeks potential correlations between those changes and alterations in neurotransmitter levels and behavioral novelty responses. Amphetamine 61-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-174 7784961-3 1995 This study illustrates how a 2 week, twice daily 7.5 mg/kg d-amphetamine or saline regimen alters rat brain regional expression of transcription factor genes, including c-fos, fos-B, jun-B, c-jun, and zif 268, and seeks potential correlations between those changes and alterations in neurotransmitter levels and behavioral novelty responses. Sodium Chloride 76-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-174 7757479-1 1995 Previous studies indicated that sexual behavior in female rats primed with estradiol and progesterone induced expression of the immediate early gene (IEG) c-Fos in various brain areas rich in estradiol receptors, including the medial preoptic area (MPA), the medial amygdala (AMe), and the ventromedial nucleus of the hypothalamus (VMN), and to a lesser extent areas with low densities of estradiol receptors, such as the caudate nucleus, the dentate gyrus and the cingulate cortex. Estradiol 75-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-160 7757479-1 1995 Previous studies indicated that sexual behavior in female rats primed with estradiol and progesterone induced expression of the immediate early gene (IEG) c-Fos in various brain areas rich in estradiol receptors, including the medial preoptic area (MPA), the medial amygdala (AMe), and the ventromedial nucleus of the hypothalamus (VMN), and to a lesser extent areas with low densities of estradiol receptors, such as the caudate nucleus, the dentate gyrus and the cingulate cortex. Progesterone 89-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-160 7864828-11 1995 Our data indicate that transcriptional activation of c-fos through its promoter by butyrate resulted in increased Fos protein expression. Butyrates 83-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 7777163-0 1995 c-fos antisense oligodeoxynucleotide increases formalin-induced nociception and regulates preprodynorphin expression. Formaldehyde 47-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 7777163-1 1995 Rats, receiving an intrathecal pretreatment of oligodeoxynucleotide complementary to c-fos mRNA (antisense), showed no increases in Fos protein or preprodynorphin messenger RNA in the outer laminae of the lumbar spinal cord when challenged 4 h later with a 50 microliters intraplantar injection of 5% formalin. Oligodeoxyribonucleotides 47-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 7777163-2 1995 Animals pretreated with saline or sense oligodeoxynucleotide showed marked increases in Fos protein (2 h after formalin challenge) and preprodynorphin mRNA (20 h after formalin challenge) in the lumbar region of the cord ipsilateral to the side of the injection. Sodium Chloride 24-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 7777163-2 1995 Animals pretreated with saline or sense oligodeoxynucleotide showed marked increases in Fos protein (2 h after formalin challenge) and preprodynorphin mRNA (20 h after formalin challenge) in the lumbar region of the cord ipsilateral to the side of the injection. Oligodeoxyribonucleotides 40-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 7777163-4 1995 These observations could be interpreted as representing a sequence of events beginning with the formalin-induced increase in the transcription factor Fos, which in turn increases the synthesis of preprodynorphin messenger RNA resulting in the production of the dynorphin opioid peptides which then exert a modulatory antinociceptive action. Formaldehyde 96-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-153 7840325-1 1995 We recently demonstrated that a meal induces c-fos immunoreactivity in the dorsal motor nucleus of the vagus (DMV), the nucleus of the tractus solitarius (NTS), and the area postrema (AP) of the rat brain stem. (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 110-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 7711171-13 1995 Both progesterone and 8-br-cAMP, which block insulin-dependent GC mitosis, also inhibited the expression of c-fos and c-jun. 8-Bromo Cyclic Adenosine Monophosphate 22-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 7536097-2 1995 Three hours after intraplantar carrageenin (6 mg/150 microliters of saline) Fos-like immunoreactivity (Fos-LI) was mainly observed in L4 and L5 segments of the dorsal horn. Carrageenan 31-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 7536097-2 1995 Three hours after intraplantar carrageenin (6 mg/150 microliters of saline) Fos-like immunoreactivity (Fos-LI) was mainly observed in L4 and L5 segments of the dorsal horn. Carrageenan 31-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-106 7536097-2 1995 Three hours after intraplantar carrageenin (6 mg/150 microliters of saline) Fos-like immunoreactivity (Fos-LI) was mainly observed in L4 and L5 segments of the dorsal horn. Sodium Chloride 68-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 7536097-2 1995 Three hours after intraplantar carrageenin (6 mg/150 microliters of saline) Fos-like immunoreactivity (Fos-LI) was mainly observed in L4 and L5 segments of the dorsal horn. Sodium Chloride 68-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-106 7536097-5 1995 We have studied the effect of systemic administration of a nitric oxide synthase inhibitor, NG-nitro-L-arginine methyl ester (L-NAME) on carrageenin evoked c-Fos expression and thus the contribution of nitric oxide to this expression. nitric 59-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 7536097-5 1995 We have studied the effect of systemic administration of a nitric oxide synthase inhibitor, NG-nitro-L-arginine methyl ester (L-NAME) on carrageenin evoked c-Fos expression and thus the contribution of nitric oxide to this expression. NG-Nitroarginine Methyl Ester 92-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 7536097-5 1995 We have studied the effect of systemic administration of a nitric oxide synthase inhibitor, NG-nitro-L-arginine methyl ester (L-NAME) on carrageenin evoked c-Fos expression and thus the contribution of nitric oxide to this expression. NG-Nitroarginine Methyl Ester 126-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 7536097-5 1995 We have studied the effect of systemic administration of a nitric oxide synthase inhibitor, NG-nitro-L-arginine methyl ester (L-NAME) on carrageenin evoked c-Fos expression and thus the contribution of nitric oxide to this expression. Carrageenan 137-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 7536097-5 1995 We have studied the effect of systemic administration of a nitric oxide synthase inhibitor, NG-nitro-L-arginine methyl ester (L-NAME) on carrageenin evoked c-Fos expression and thus the contribution of nitric oxide to this expression. Nitric Oxide 59-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 7536097-18 1995 ), the inactive isomer of L-NAME, produced a weak reduction of the number of superficial laminae Fos-LI neurones (26 +/- 8% reduction, P<0.05), without influencing the deep Fos-LI neurones (5 +/- 8% enhancement) or the oedema.8. NG-Nitroarginine Methyl Ester 26-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 7536097-22 1995 There is a strong correlation between the reduction of the number of Fos-LI neurones and the oedema by L-NAME, clearly demonstrating a predominant role of peripheral NO in the development of one of the signs of carrageenin inflammation. Carrageenan 211-222 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 8882576-4 1995 In neuronal cell cultures, the NMDA (0.1 microM)-induced c-fos mRNA expression on culture day 18 in vitro was higher in P77PMC than Wistar rats (P < 0.05). N-Methylaspartate 31-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 8535862-0 1995 Roles of dopamine D1 receptors in striatal fos protein induction associated with methamphetamine behavioral sensitization in rats. Methamphetamine 81-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 8535862-2 1995 Intraperitoneal administration of 5.0 mg/kg methamphetamine produced a significant increase in Fos-immunoreactive cells in the medial striatum. Methamphetamine 44-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 8535862-6 1995 These results suggest that dopamine D1 receptor-mediated mechanisms are involved in the striatal Fos protein induction associated with behavioral sensitization following exposure to methamphetamine. Methamphetamine 182-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 8777434-5 1995 All trans-retinoic acid (RA) abolished the transformation of the 43C line and TPA-treated RELcJ1 cells, suggesting that RA could decrease AP1 activity in these cells despite c-fos or c-jun overexpression. Tretinoin 4-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 174-179 7707864-6 1995 Intraperitoneal injection of MK-801 (3 mg/kg), a non-competitive NMDA receptor antagonist, suppressed the expression of c-fos mRNA after cortical ablation. Dizocilpine Maleate 29-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 8589759-5 1995 When myocytes were exposed to nonlethal concentrations of H2O2, c-fos and c-jun mRNAs were rapidly induced, reaching peak values at 30-60 min. Hydrogen Peroxide 58-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 7760370-1 1995 We have studied the effects of pressure and volume overload as well as of norepinephrine (NE) alone and in combination on the expression of the proto-oncogenes c-fos and c-myc in isolated perfused working rat hearts. Norepinephrine 74-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 8748612-3 1995 High doses of MK 801 or CPP infused into the SN produced intense contralateral turning per-se but induced only sparse c-fos expression in the lesioned CPu. Dizocilpine Maleate 14-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 7823174-9 1995 Diazepam also produced dose-related decreases in conditioned stress-induced c-fos expression in all but one structure, the effects being statistically significant in 38 of 60 sampled structures. Diazepam 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 7823174-10 1995 Diazepam dose dependently increased fear-induced c-fos expression in the central nucleus of the amygdala. Diazepam 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 7823174-13 1995 The extent to which diazepam decreased conditioned stress-induced c-fos expression was unrelated to previous estimates of benzodiazepine receptor density in the sampled structures. Diazepam 20-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 7823175-0 1995 Alcohol selectively attenuates stress-induced c-fos expression in rat hippocampus. Alcohols 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 7823175-6 1995 An intraperitoneal injection of 2 g/kg alcohol prior to stress decreased c-Fos expression in several but not all of these structures. Alcohols 39-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 7823175-7 1995 In particular, alcohol strongly attenuated the stress-induced expression of c-Fos in hippocampus and cingulate cortex. Alcohols 15-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 7823175-8 1995 Using slot-blot hybridization, significant induction of c-fos mRNA after restraint stress was demonstrated both in hippocampus and cortex, but prior alcohol exposure specifically attenuated c-fos induction only in the hippocampus. Alcohols 149-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 190-195 7533872-2 1995 In isolated rat hepatocytes angiotensin II and phorbol 12-myristate 13-acetate (PMA) induce the expression of c-fos. Tetradecanoylphorbol Acetate 47-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 7533872-2 1995 In isolated rat hepatocytes angiotensin II and phorbol 12-myristate 13-acetate (PMA) induce the expression of c-fos. Tetradecanoylphorbol Acetate 80-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 7533872-6 1995 Inhibitors of serine-threonine protein phosphatases, such as okadaic acid, microcystin LR and calyculin also induce c-fos expression. Okadaic Acid 61-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 7533872-6 1995 Inhibitors of serine-threonine protein phosphatases, such as okadaic acid, microcystin LR and calyculin also induce c-fos expression. cyanoginosin LR 75-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 7533872-6 1995 Inhibitors of serine-threonine protein phosphatases, such as okadaic acid, microcystin LR and calyculin also induce c-fos expression. calyculin A 94-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 8527000-6 1995 In the striata of 6-OHDA-lesioned rats, PENK gene is upregulated, PDYN gene is down-regulated and the induction of c-fos gene by D2 receptor antagonists is abolished, whereas selective D1 receptor agonists induce c-fos gene, which does not occur in non-lesioned rats. Oxidopamine 18-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 8527000-6 1995 In the striata of 6-OHDA-lesioned rats, PENK gene is upregulated, PDYN gene is down-regulated and the induction of c-fos gene by D2 receptor antagonists is abolished, whereas selective D1 receptor agonists induce c-fos gene, which does not occur in non-lesioned rats. Oxidopamine 18-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-218 8748612-0 1995 Intranigral injections of glutamate antagonists modulate dopamine D1-mediated turning behavior and striatal c-fos expression. Glutamic Acid 26-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 8748612-2 1995 Local infusion into the SN of the 6-OHDA lesioned side of NMDA glutamate antagonists MK 801 and CPP or the AMPA antagonist NBQX at doses inducing none or minimal behavioral effects, significantly increased the turning behavior and the expression of c-fos induced, in the lesioned caudate-putamen (CPu), by a parenteral administration of SKF 38393. Oxidopamine 34-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 249-254 8748612-2 1995 Local infusion into the SN of the 6-OHDA lesioned side of NMDA glutamate antagonists MK 801 and CPP or the AMPA antagonist NBQX at doses inducing none or minimal behavioral effects, significantly increased the turning behavior and the expression of c-fos induced, in the lesioned caudate-putamen (CPu), by a parenteral administration of SKF 38393. NMDA glutamate 58-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 249-254 8748612-2 1995 Local infusion into the SN of the 6-OHDA lesioned side of NMDA glutamate antagonists MK 801 and CPP or the AMPA antagonist NBQX at doses inducing none or minimal behavioral effects, significantly increased the turning behavior and the expression of c-fos induced, in the lesioned caudate-putamen (CPu), by a parenteral administration of SKF 38393. Dizocilpine Maleate 85-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 249-254 8748612-2 1995 Local infusion into the SN of the 6-OHDA lesioned side of NMDA glutamate antagonists MK 801 and CPP or the AMPA antagonist NBQX at doses inducing none or minimal behavioral effects, significantly increased the turning behavior and the expression of c-fos induced, in the lesioned caudate-putamen (CPu), by a parenteral administration of SKF 38393. 2,3-dioxo-6-nitro-7-sulfamoylbenzo(f)quinoxaline 123-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 249-254 8777434-7 1995 The spontaneous transformation of the c-fos-transfected REL cells was associated with the appearance of c-fos/AP1 complexes binding TRE, suggesting that c-fos/AP1 complexes are involved in the antitransforming mechanism of quercetin. Quercetin 223-232 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 8777434-7 1995 The spontaneous transformation of the c-fos-transfected REL cells was associated with the appearance of c-fos/AP1 complexes binding TRE, suggesting that c-fos/AP1 complexes are involved in the antitransforming mechanism of quercetin. Quercetin 223-232 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 8777434-7 1995 The spontaneous transformation of the c-fos-transfected REL cells was associated with the appearance of c-fos/AP1 complexes binding TRE, suggesting that c-fos/AP1 complexes are involved in the antitransforming mechanism of quercetin. Quercetin 223-232 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 7796933-6 1995 Consistently, ionomycin plus low doses of TPA solely reproduced the potent effect of TRH on c-fos transcripts. Ionomycin 14-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 7796933-6 1995 Consistently, ionomycin plus low doses of TPA solely reproduced the potent effect of TRH on c-fos transcripts. Tetradecanoylphorbol Acetate 42-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 7796933-10 1995 The overall study also reveals that among the agonists tested, the dihydropyridine Bay K 8644 and forskolin only were capable to induce a long-lasting stimulation of c-fos and jun B mRNA levels, concomitant to increased levels of PRL transcripts, as does TRH. Colforsin 98-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-171 7796933-7 1995 Data collected from TRH and TPA down-regulated cells indicated that TRH probably recruits TPA-dependent PKC isoforms for stimulating c-fos but not jun B transcripts. Tetradecanoylphorbol Acetate 28-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 7796933-7 1995 Data collected from TRH and TPA down-regulated cells indicated that TRH probably recruits TPA-dependent PKC isoforms for stimulating c-fos but not jun B transcripts. Tetradecanoylphorbol Acetate 90-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 7796933-10 1995 The overall study also reveals that among the agonists tested, the dihydropyridine Bay K 8644 and forskolin only were capable to induce a long-lasting stimulation of c-fos and jun B mRNA levels, concomitant to increased levels of PRL transcripts, as does TRH. 1,4-dihydropyridine 67-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-171 7787765-0 1995 Methamphetamine-induced nuclear c-Fos in rat brain regions. Methamphetamine 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 7663880-0 1995 A sex comparison of increments in FOS immunoreactivity in forebrain neurons of gonadectomized, testosterone-treated rats after mounting an estrous female. Testosterone 95-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 7663880-1 1995 The protein product of the immediate-early gene, c-fos, was visualized immunocytochemically in forebrain neurons of gonadectomized male and female rats which were injected daily with testosterone propionate (TP) and either tested for mounting directed toward a sexually receptive female or left alone in a test arena. Testosterone Propionate 183-206 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 7663880-1 1995 The protein product of the immediate-early gene, c-fos, was visualized immunocytochemically in forebrain neurons of gonadectomized male and female rats which were injected daily with testosterone propionate (TP) and either tested for mounting directed toward a sexually receptive female or left alone in a test arena. Testosterone Propionate 208-210 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 7787765-1 1995 To explore the possible robust changes in neuronal activity in dopamine-poor brain regions after an indirect dopamine agonist, methamphetamine, we have investigated its effects on c-fos expression in rat brain using immunocytochemistry of c-Fos. Methamphetamine 127-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 180-185 7787765-2 1995 Intraperitoneal injection of methamphetamine (1.6-4.8 mg/kg), but not of saline, induced a widespread nuclear c-Fos-like immunoreactivity in the pyriform cortex and olfactory tubercle with greatest density followed by the II-VI layers of the neocortex, amygdala, hypothalamus, thalamus, nucleus accumbens and striatum. Methamphetamine 29-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 7709341-0 1995 Cocaine and d-amphetamine increase c-fos expression in the rat cerebellum. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 8896075-2 1995 We analyze the effects produced by the simultaneous administration of adenosinetriphosphate (fosfobion) (FOS) and Isoproterenol on the changes in the body weight/heart weight ratio, and on the biochemical changes of cardiac metabolism. Adenosine Triphosphate 70-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-108 8896075-2 1995 We analyze the effects produced by the simultaneous administration of adenosinetriphosphate (fosfobion) (FOS) and Isoproterenol on the changes in the body weight/heart weight ratio, and on the biochemical changes of cardiac metabolism. Adenosine Triphosphate 93-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-108 8896075-6 1995 Significant changes were found in the myocardium lactate that decreased by 26% under ISO influence, in comparison with normal values, and that decreased by 90% when FOS and ISO were administered together. Lactic Acid 49-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-168 7709341-0 1995 Cocaine and d-amphetamine increase c-fos expression in the rat cerebellum. Dextroamphetamine 12-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 7709341-2 1995 In the present study, immunohistochemical techniques were used to assess the pattern of c-fos expression in the cerebellum produced by d-amphetamine or cocaine. Dextroamphetamine 135-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 7709341-2 1995 In the present study, immunohistochemical techniques were used to assess the pattern of c-fos expression in the cerebellum produced by d-amphetamine or cocaine. Cocaine 152-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 7709341-5 1995 Dose-dependent increases in Fos-like immunoreactivity were elicited by d-amphetamine and cocaine. Dextroamphetamine 71-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 7709341-5 1995 Dose-dependent increases in Fos-like immunoreactivity were elicited by d-amphetamine and cocaine. Cocaine 89-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 7709341-8 1995 In addition, a homogeneous pattern of Fos-like immunoreactive nuclei, of sparse density, was also found near the pial surface of the molecular layer following d-amphetamine but not cocaine. Dextroamphetamine 159-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 7888504-0 1994 A reduced proportion of luteinizing hormone (LH)-releasing hormone neurons express Fos protein during the preovulatory or steroid-induced LH surge in middle-aged rats. Steroids 122-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 7718144-8 1994 C-fos studies suggested that association between gustatory CS and visceral US takes place in the PBN. Cesium 59-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 7888504-4 1994 The mean proportion of LHRH neurons containing immunoreactive Fos was higher in the brains of young compared to middle-aged females in association with both the preovulatory (p < 0.01) and the steroid-induced LH surge (p < 0.001). Steroids 196-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 7888504-4 1994 The mean proportion of LHRH neurons containing immunoreactive Fos was higher in the brains of young compared to middle-aged females in association with both the preovulatory (p < 0.01) and the steroid-induced LH surge (p < 0.001). Luteinizing Hormone 23-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 7988435-13 1994 a convergence of TPA, FSH, and cAMP mediated signals in prepubertal Sertoli cells may occur with the induction of specific AP-1 site binding complex(es) containing jun and fos proteins. Tetradecanoylphorbol Acetate 17-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-175 7898306-7 1994 Lithium in combination with other inducers caused delayed increases in both AP-1 binding activity and c-jun, c-fos and fra-1 gene expression. Lithium 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 7829168-9 1994 The induction of c-fos was completely blocked or reduced by treatment with MK-801. Dizocilpine Maleate 75-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 7527386-3 1994 The Stat91 pathway regulated c-fos gene transcription involves activation by tyrosine phosphorylation of the DNA binding factor SIF (sis-inducing factor) in the cytoplasm, its nuclear translocation, and interaction with the regulatory element SIE (sis-inducing element). Tyrosine 77-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 7996173-0 1994 Differential effects of morphine on noxious stimulus-evoked fos-like immunoreactivity in subpopulations of spinoparabrachial neurons. Morphine 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 7996173-1 1994 In previous studies we reported that although morphine dose dependently inhibits noxious stimulus-evoked expression of the c-fos proto-oncogene in the rat spinal cord, morphine was without effect in certain populations of presumed nociresponsive neurons, even under conditions of complete behavioral analgesia. Morphine 46-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 7996173-2 1994 To determine whether the neurons that continue to express the c-fos gene include projection neurons, we evaluated the effect of morphine on noxious stimulus-evoked c-fos expression in spinoparabrachial neurons retrogradely labeled with Fluoro-gold. Morphine 128-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-169 7996173-3 1994 In the formalin test, we found that morphine analgesia was associated with a significant reduction in the number of Fos-like-immunoreactive spinoparabrachial projection neurons in the lateral reticulated area of the neck of the dorsal horn. Formaldehyde 7-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 7996173-3 1994 In the formalin test, we found that morphine analgesia was associated with a significant reduction in the number of Fos-like-immunoreactive spinoparabrachial projection neurons in the lateral reticulated area of the neck of the dorsal horn. Morphine 36-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 7996173-5 1994 These results indicate that under conditions of morphine analgesia two distinct populations of spinoparabrachial neurons can be recognized on the basis of their expression of the c-fos gene in response to noxious stimulation. Morphine 48-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-184 7996173-6 1994 Since the expression of the c-fos gene has been correlated with neuronal activity, these data suggest that activity, and central transmission of nociceptive information, persists in certain nociresponsive projection neurons during morphine analgesia. Morphine 231-239 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 7898680-5 1994 In the remainder of the spinal cord, between 21 and 35% of the c-fos-immunoreactive cells were GABA- or glycine-immunoreactive, and the majority of these neurons contained both types of immunoreactivity. Glycine 104-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 7898680-7 1994 It is known that some of the cells which produce c-fos in response to noxious stimulation are projection neurons, with axons ascending to the brainstem or thalamus, however, because of the large number of c-fos-immunoreactive cells in the dorsal horn, it is likely that many are interneurons, and some of these are probably excitatory cells which use glutamate as a transmitter. Glutamic Acid 351-360 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 7970716-0 1994 Retinoic acid suppresses polyoma virus transformation by inhibiting transcription of the c-fos proto-oncogene. Tretinoin 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 7970716-3 1994 In this report we present the results of experiments that demonstrate that retinoic acid does indeed inhibit transcriptional transactivation of the c-fos promoter by polyoma virus, as well as by calf serum and purified serum growth factors. Tretinoin 75-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 7970716-5 1994 Restoration of c-fos expression, even in the presence of retinoic acid, restored transformation, indicating that retinoic acid inhibition of c-fos expression is sufficient to explain the retinoid suppression of transformation. Tretinoin 113-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 7970716-5 1994 Restoration of c-fos expression, even in the presence of retinoic acid, restored transformation, indicating that retinoic acid inhibition of c-fos expression is sufficient to explain the retinoid suppression of transformation. Tretinoin 113-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 7970716-5 1994 Restoration of c-fos expression, even in the presence of retinoic acid, restored transformation, indicating that retinoic acid inhibition of c-fos expression is sufficient to explain the retinoid suppression of transformation. Retinoids 187-195 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 7970716-5 1994 Restoration of c-fos expression, even in the presence of retinoic acid, restored transformation, indicating that retinoic acid inhibition of c-fos expression is sufficient to explain the retinoid suppression of transformation. Retinoids 187-195 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 7970716-6 1994 These results identify the c-fos proto-oncogene as a key nuclear target for mT-dependent transformation and show that the anticarcinogenic properties of retinoic acid can be brought about by inhibiting c-fos expression. Tretinoin 153-166 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 7970716-6 1994 These results identify the c-fos proto-oncogene as a key nuclear target for mT-dependent transformation and show that the anticarcinogenic properties of retinoic acid can be brought about by inhibiting c-fos expression. Tretinoin 153-166 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 202-207 7886620-0 1994 Differential effect of MK 801 and scopolamine on c-fos expression induced by L-dopa in the striatum of 6-hydroxydopamine lesioned rats. Dizocilpine Maleate 23-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 7886620-0 1994 Differential effect of MK 801 and scopolamine on c-fos expression induced by L-dopa in the striatum of 6-hydroxydopamine lesioned rats. Scopolamine 34-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 7886620-0 1994 Differential effect of MK 801 and scopolamine on c-fos expression induced by L-dopa in the striatum of 6-hydroxydopamine lesioned rats. Levodopa 77-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 7886620-0 1994 Differential effect of MK 801 and scopolamine on c-fos expression induced by L-dopa in the striatum of 6-hydroxydopamine lesioned rats. Oxidopamine 103-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 7886620-1 1994 In rats with a unilateral 6-hydroxydopamine lesion of the dopaminergic nigro-striatal pathway, striatal D1-receptor-stimulated c-fos expression and turning behavior are positively modulated by D2 receptor stimulation and by blockade of N-methyl-D-aspartate (NMDA) or muscarinic receptors. Oxidopamine 26-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 7886620-1 1994 In rats with a unilateral 6-hydroxydopamine lesion of the dopaminergic nigro-striatal pathway, striatal D1-receptor-stimulated c-fos expression and turning behavior are positively modulated by D2 receptor stimulation and by blockade of N-methyl-D-aspartate (NMDA) or muscarinic receptors. N-Methylaspartate 236-256 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 7886620-1 1994 In rats with a unilateral 6-hydroxydopamine lesion of the dopaminergic nigro-striatal pathway, striatal D1-receptor-stimulated c-fos expression and turning behavior are positively modulated by D2 receptor stimulation and by blockade of N-methyl-D-aspartate (NMDA) or muscarinic receptors. N-Methylaspartate 258-262 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 7886620-2 1994 Combined D1/D2 receptor stimulation by L-dopa activates c-fos in a manner not additive with muscarinic receptor blockade by scopolamine. Levodopa 39-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 7886620-3 1994 On the other hand, blockade of NMDA receptors by MK 801 reduced c-fos expression induced by L-dopa while, depending on the dose of L-dopa, differentially affecting contralateral turning behavior. Dizocilpine Maleate 49-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 7886620-3 1994 On the other hand, blockade of NMDA receptors by MK 801 reduced c-fos expression induced by L-dopa while, depending on the dose of L-dopa, differentially affecting contralateral turning behavior. Levodopa 92-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 7886627-1 1994 The role of N-methyl-D-aspartate (NMDA) excitatory amino acid receptors in D-amphetamine (AMPH)-induced behavioral changes and increased expression of the nuclear transcription factors, c-fos and zif/268, and preprodynorphin (PPD) mRNA in various regions of rat forebrain was investigated with quantitative in situ hybridization histochemistry. Dextroamphetamine 75-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 186-191 7886627-1 1994 The role of N-methyl-D-aspartate (NMDA) excitatory amino acid receptors in D-amphetamine (AMPH)-induced behavioral changes and increased expression of the nuclear transcription factors, c-fos and zif/268, and preprodynorphin (PPD) mRNA in various regions of rat forebrain was investigated with quantitative in situ hybridization histochemistry. Dextroamphetamine 90-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 186-191 7882006-0 1994 NMDA receptor-mediated expression of Fos protein in the rat striatum following methamphetamine administration: relation to behavioral sensitization. Methamphetamine 79-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 7882006-2 1994 A single administration of 1.0 and 5.0 mg/kg methamphetamine resulted in a dose-dependent increase in Fos-immunoreactive cells in the medial striatum. Methamphetamine 45-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 7882006-4 1994 Pretreatment with 1.0 mg/kg MK-801 completely prevented both the expression of striatal Fos protein and the development of acute behavioral sensitization following a single injection of 5.0 mg/kg methamphetamine. Dizocilpine Maleate 28-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 7882006-4 1994 Pretreatment with 1.0 mg/kg MK-801 completely prevented both the expression of striatal Fos protein and the development of acute behavioral sensitization following a single injection of 5.0 mg/kg methamphetamine. Methamphetamine 196-211 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 7882006-5 1994 These results suggest that NMDA receptor-mediated mechanisms contribute to the expression of striatal Fos protein associated with behavioral sensitization that follows exposure to methamphetamine. Methamphetamine 180-195 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 7882012-0 1994 Transection of corticostriatal afferents abolishes the hyperexpression of Fos and counteracts the development of rotational overcompensation induced by intrastriatal dopamine-rich grafts when challenged with amphetamine. Amphetamine 208-219 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 7882012-1 1994 The present study was carried out to test whether the abnormally high striatal Fos activation induced by amphetamine and the overcompensation of amphetamine-induced rotation in 6-hydroxydopamine-lesioned rats receiving transplants of fetal nigral neurons can be reduced by a lesion of the corticostriatal projection. Amphetamine 105-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 7999013-1 1994 Basal and stimulated activity of the c-fos promoter is reduced by triiodothyronine (T3) and retinoic acid (RA) in GH1 cells. Triiodothyronine 66-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 7999013-1 1994 Basal and stimulated activity of the c-fos promoter is reduced by triiodothyronine (T3) and retinoic acid (RA) in GH1 cells. Triiodothyronine 84-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 7999013-1 1994 Basal and stimulated activity of the c-fos promoter is reduced by triiodothyronine (T3) and retinoic acid (RA) in GH1 cells. Tretinoin 92-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 7999013-1 1994 Basal and stimulated activity of the c-fos promoter is reduced by triiodothyronine (T3) and retinoic acid (RA) in GH1 cells. Tretinoin 107-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 7860779-7 1994 Enflurane alone induced c-fos expression in the same brain area; however, the number of Fos-positive cells and double-labeled cells were decreased two- to fivefold and three- to eightfold, respectively, compared with the abdominal surgery groups. Enflurane 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 7887109-2 1994 In this study, the expression of c-fos protein (c-Fos) following lignocaine-induced convulsions was examined and compared with that following convulsions induced by non-anesthetic convulsants, such as pentylenetetrazol, kainic acid and electroconvulsive shocks, in rat brain. Lidocaine 65-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 7887109-2 1994 In this study, the expression of c-fos protein (c-Fos) following lignocaine-induced convulsions was examined and compared with that following convulsions induced by non-anesthetic convulsants, such as pentylenetetrazol, kainic acid and electroconvulsive shocks, in rat brain. Lidocaine 65-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 7887109-2 1994 In this study, the expression of c-fos protein (c-Fos) following lignocaine-induced convulsions was examined and compared with that following convulsions induced by non-anesthetic convulsants, such as pentylenetetrazol, kainic acid and electroconvulsive shocks, in rat brain. Pentylenetetrazole 201-218 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 7887109-2 1994 In this study, the expression of c-fos protein (c-Fos) following lignocaine-induced convulsions was examined and compared with that following convulsions induced by non-anesthetic convulsants, such as pentylenetetrazol, kainic acid and electroconvulsive shocks, in rat brain. Kainic Acid 220-231 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 7887109-2 1994 In this study, the expression of c-fos protein (c-Fos) following lignocaine-induced convulsions was examined and compared with that following convulsions induced by non-anesthetic convulsants, such as pentylenetetrazol, kainic acid and electroconvulsive shocks, in rat brain. Kainic Acid 220-231 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 7700008-2 1994 Neurons in the brain responsive to the subcutaneous infusion of isoproterenol were identified using an antibody to Fos, the protein product of c-fos which is now used extensively as a marker of activated neurons. Isoproterenol 64-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-118 7700008-2 1994 Neurons in the brain responsive to the subcutaneous infusion of isoproterenol were identified using an antibody to Fos, the protein product of c-fos which is now used extensively as a marker of activated neurons. Isoproterenol 64-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-148 7898680-3 1994 In the superficial dorsal horn (laminae I and II) and dorsal white matter, between 14 and 20% of c-fos-immunoreactive neurons were GABA-immunoreactive, and some of these were also glycine-immunoreactive. gamma-Aminobutyric Acid 131-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 7898680-5 1994 In the remainder of the spinal cord, between 21 and 35% of the c-fos-immunoreactive cells were GABA- or glycine-immunoreactive, and the majority of these neurons contained both types of immunoreactivity. gamma-Aminobutyric Acid 95-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 7878091-1 1994 The distribution of evoked expression of the proto-oncogene c-fos was immunohistochemically examined in the parabrachial nucleus (PBN) of water-deprived rats after free ingestion of palatable liquids, after intra-oral infusion of aversive taste solutions including various bitter substances, and after an intraperitoneal injection of LiCl. Water 138-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-65 7878091-2 1994 C-fos immunoreactive neurons (c-fos neurons) were densely packed in the external lateral subnucleus (els), external medial subnucleus (ems), dorsal lateral subnucleus (dls), central lateral subnucleus (cls), and the central medial subnucleus (cms) depending on the kind of stimulation. N-[(2S,3S,4R)-3,4-dihydroxy-8-oxo-8-[(4-pentylphenyl)amino]-1-{[(2S,3R,4S,5R,6R)-3,4,5-trihydroxy-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yl]oxy}octan-2-yl]hexacosanamide 101-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 7878091-2 1994 C-fos immunoreactive neurons (c-fos neurons) were densely packed in the external lateral subnucleus (els), external medial subnucleus (ems), dorsal lateral subnucleus (dls), central lateral subnucleus (cls), and the central medial subnucleus (cms) depending on the kind of stimulation. N-[(2S,3S,4R)-3,4-dihydroxy-8-oxo-8-[(4-pentylphenyl)amino]-1-{[(2S,3R,4S,5R,6R)-3,4,5-trihydroxy-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yl]oxy}octan-2-yl]hexacosanamide 101-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 7878091-2 1994 C-fos immunoreactive neurons (c-fos neurons) were densely packed in the external lateral subnucleus (els), external medial subnucleus (ems), dorsal lateral subnucleus (dls), central lateral subnucleus (cls), and the central medial subnucleus (cms) depending on the kind of stimulation. dls 168-171 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 7878091-2 1994 C-fos immunoreactive neurons (c-fos neurons) were densely packed in the external lateral subnucleus (els), external medial subnucleus (ems), dorsal lateral subnucleus (dls), central lateral subnucleus (cls), and the central medial subnucleus (cms) depending on the kind of stimulation. dls 168-171 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 7878091-2 1994 C-fos immunoreactive neurons (c-fos neurons) were densely packed in the external lateral subnucleus (els), external medial subnucleus (ems), dorsal lateral subnucleus (dls), central lateral subnucleus (cls), and the central medial subnucleus (cms) depending on the kind of stimulation. calusterone 202-205 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 7878091-2 1994 C-fos immunoreactive neurons (c-fos neurons) were densely packed in the external lateral subnucleus (els), external medial subnucleus (ems), dorsal lateral subnucleus (dls), central lateral subnucleus (cls), and the central medial subnucleus (cms) depending on the kind of stimulation. calusterone 202-205 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 7891895-0 1994 Induction of Fos-like immunoreactivity in the rat hypothalamus by an endogenous feeding suppressant (2-buten-4-olide). butenolide 101-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 7891895-1 1994 Central influence of 2-buten-4-olide (2-B4O), an endogenous feeding suppressant, was studied through immunohistochemical detection of Fos expression in the rat hypothalamus. butenolide 21-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-137 7891895-1 1994 Central influence of 2-buten-4-olide (2-B4O), an endogenous feeding suppressant, was studied through immunohistochemical detection of Fos expression in the rat hypothalamus. butenolide 38-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-137 7891895-5 1994 Immunohistochemical analysis revealed that injection of 2-B4O induced Fos-like immunoreactivity significantly in the paraventricular, dorsomedial and arcuate nuclei and the periventricular and lateral hypothalamic areas. butenolide 56-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 7874510-6 1994 Nutrients effective at decreasing food intake (carbohydrate and fat) produced significant increases in Fos-like immunopositive cells in the NTS. Carbohydrates 47-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-106 7654338-6 1994 Indeed, cotreatment of thymocytes with the nonspecific serine protease inhibitor phenylmethylsulphonyl fluoride was able to partially protect the stability of the DNA-binding proteins and alter the expression of the c-fos and c-jun genes but did not inhibit apoptosis. Phenylmethylsulfonyl Fluoride 81-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 216-221 7988435-13 1994 a convergence of TPA, FSH, and cAMP mediated signals in prepubertal Sertoli cells may occur with the induction of specific AP-1 site binding complex(es) containing jun and fos proteins. Cyclic AMP 31-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-175 7900542-6 1994 These results seem to contradict the previous reports of no c-fos induction in rats this young by demonstrating a functional c-fos translational mechanism by GD-21. gd-21 158-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 7858563-9 1994 Amiloride, an inhibitor of Na+/H+ exchange, decreased rates of protein synthesis in a concentration-dependent manner (12.5, 25, 50, 100 microM) but did not change cAMP content (5.25 +/- 0.11 pmol/mg protein) or expression of c-fos mRNA. Amiloride 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 225-230 7936645-13 1994 Superfusion of rat skin with c-fos antisense oligodeoxynucleotides inhibited the increase in c-Fos immunolabeled epidermal cells 1.5 h after single u.v. Oligodeoxyribonucleotides 45-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 7936645-13 1994 Superfusion of rat skin with c-fos antisense oligodeoxynucleotides inhibited the increase in c-Fos immunolabeled epidermal cells 1.5 h after single u.v. Oligodeoxyribonucleotides 45-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 7855736-0 1994 Amphetamine- and cocaine-induced fos in the rat striatum depends on D2 dopamine receptor activation. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 7931352-1 1994 Administration of neuroleptics such as haloperidol to rats is known to induce the expression of the immediate-early genes (IEGs) c-fos and zif/268 in striatal neurones. Haloperidol 39-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 7882899-6 1994 cdc2 p34 and the c-fos 28 kDa protein were found in estradiol-treated rat uteri and were not detected in uteri of control and ICI 182,780-treated animals; whereas Cyclin B1 was absent in uteri from control and estradiol-treated ovariectomized animals. Estradiol 52-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 7882899-7 1994 ICI 182,780 administered to estradiol-treated ovariectomized rats blocked the induction of cdc2 p34 and the c-fos 28 kDa protein in the uterus. Estradiol 28-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 7855736-4 1994 Eticlopride treatment (0.5 mg/kg) caused Fos expression by itself, but also decreased Fos expression in the central striatum due to amphetamine (5.0 mg/kg) or cocaine (40 mg/kg) by 90% and 85%, respectively. Amphetamine 132-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 7855736-5 1994 In striatonigral neurons, identified by labeling with the retrograde tracer Fluorogold iontophoresed into the substantia nigra pars reticulata, the blockade of stimulant-induced Fos-like immunofluorescence by eticlopride was nearly complete, with decreases of 98% for amphetamine and 94% for cocaine. eticlopride 209-220 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 178-181 7855736-5 1994 In striatonigral neurons, identified by labeling with the retrograde tracer Fluorogold iontophoresed into the substantia nigra pars reticulata, the blockade of stimulant-induced Fos-like immunofluorescence by eticlopride was nearly complete, with decreases of 98% for amphetamine and 94% for cocaine. Amphetamine 268-279 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 178-181 7855736-5 1994 In striatonigral neurons, identified by labeling with the retrograde tracer Fluorogold iontophoresed into the substantia nigra pars reticulata, the blockade of stimulant-induced Fos-like immunofluorescence by eticlopride was nearly complete, with decreases of 98% for amphetamine and 94% for cocaine. Cocaine 292-299 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 178-181 7855736-7 1994 We conclude that activation of both D1 and D2 receptor classes by dopamine agonists is necessary for induction of Fos in the striatonigral cells of normal rats. Dopamine 66-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 7859070-3 1994 Thus, in terms of number of cells, haemorrhage was significantly more potent than water deprivation in inducing Fos immunoreactivity in either type of neuron in the supraoptic, paraventricular and anterior commissural nuclei. Water 82-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 7855736-0 1994 Amphetamine- and cocaine-induced fos in the rat striatum depends on D2 dopamine receptor activation. Cocaine 17-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 7859070-4 1994 However, the Fos reaction of vasopressin cells in response to either stimulus was greater than that of oxytocin cells in the supraoptic and paraventricular nuclei, and in the perifornical posterior nucleus and nucleus circularis in response to water deprivation. Water 244-249 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 7855736-1 1994 Amphetamine or cocaine injection causes expression of the immediate-early gene c-fos in the striatum. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 7859070-6 1994 Finally, our analyses clearly indicated that Fos synthesis in either type of magnocellular neuron was closely linked to the reproductive state of the animal since after haemorrhage or water deprivation, the number of Fos-positive oxytocin cells in the supraoptic nucleus and Fos-positive vasopressin cells in the paraventricular nucleus was significantly less in lactating than in virgin rats. Water 184-189 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 7855736-1 1994 Amphetamine or cocaine injection causes expression of the immediate-early gene c-fos in the striatum. Cocaine 15-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 7855736-3 1994 We have investigated the involvement of D2 receptors in indirect dopamine agonist-induced striatal Fos-like immunoreactivity using the selective D2 antagonist eticlopride. Dopamine 65-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 7855736-4 1994 Eticlopride treatment (0.5 mg/kg) caused Fos expression by itself, but also decreased Fos expression in the central striatum due to amphetamine (5.0 mg/kg) or cocaine (40 mg/kg) by 90% and 85%, respectively. eticlopride 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 7855736-4 1994 Eticlopride treatment (0.5 mg/kg) caused Fos expression by itself, but also decreased Fos expression in the central striatum due to amphetamine (5.0 mg/kg) or cocaine (40 mg/kg) by 90% and 85%, respectively. eticlopride 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 7535406-0 1994 Postnatal development of nitric oxide synthase type 1 expression in the lumbar spinal cord of the rat: a comparison with the induction of c-fos in response to peripheral application of mustard oil. mustard oil 185-196 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 7859063-4 1994 Studies on c-fos like immunoreactivity after systemic D-amphetamine treatment demonstrated a wide-spread appearance of c-fos like immunoreactive neuronal nuclear profiles within the neostriatum on both the unlesioned and denervated side. Dextroamphetamine 54-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 7859063-4 1994 Studies on c-fos like immunoreactivity after systemic D-amphetamine treatment demonstrated a wide-spread appearance of c-fos like immunoreactive neuronal nuclear profiles within the neostriatum on both the unlesioned and denervated side. Dextroamphetamine 54-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 7844257-6 1994 Nitroprusside-induced isovolemic hypotension yielded a pattern of c-fos induction similar to that seen following hemorrhage, except in the area postrema and the A1 cell group, where the response was muted or lacking. Nitroprusside 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 7844257-7 1994 Phenylephrine-induced hypertension stimulated a restricted pattern of c-fos expression, largely limited to induced hypertension stimulated a restricted pattern of c-fos expression, largely limited to non-aminergic neurons, whose distribution in the NTS conformed to the termination patterns of primary baroreceptor afferents, and in the ventrolateral medulla overlapped in part with those of vagal cardiomotor and depressor neurons. Phenylephrine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 7844257-7 1994 Phenylephrine-induced hypertension stimulated a restricted pattern of c-fos expression, largely limited to induced hypertension stimulated a restricted pattern of c-fos expression, largely limited to non-aminergic neurons, whose distribution in the NTS conformed to the termination patterns of primary baroreceptor afferents, and in the ventrolateral medulla overlapped in part with those of vagal cardiomotor and depressor neurons. Phenylephrine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-168 7535406-4 1994 NOS expression in lamina II paralleled the development of c-fos expression in this lamina in response to peripheral application of mustard oil. mustard oil 131-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 7820657-0 1994 Regional suppression by water intake of c-fos expression induced by intraventricular infusions of angiotensin II. Water 24-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 7854036-2 1994 Previous studies have demonstrated that the IEGs NGFI-A (zif268) and c-fos are each rapidly induced in the caudate putamen (CP) by treatment with the indirect dopamine agonist cocaine. Dopamine 159-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 7854036-2 1994 Previous studies have demonstrated that the IEGs NGFI-A (zif268) and c-fos are each rapidly induced in the caudate putamen (CP) by treatment with the indirect dopamine agonist cocaine. Cocaine 176-183 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 7854036-8 1994 Levels of NGFI-A and c-fos were measured in the CP of rats by Northern blot analysis, which confirmed that cocaine-induced increases of NGFI-A and c-fos mRNA lasts for several hours after drug administration. Cocaine 107-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 7854036-8 1994 Levels of NGFI-A and c-fos were measured in the CP of rats by Northern blot analysis, which confirmed that cocaine-induced increases of NGFI-A and c-fos mRNA lasts for several hours after drug administration. Cocaine 107-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-152 7854036-9 1994 Immediately following this induction, however, there is a prolonged period during which a marked reduction in the relative amount of mRNA for both NGFI-A and c-fos is observed in cocaine-treated animals when compared to matched, vehicle-treated controls. Cocaine 179-186 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 7848908-5 1994 Cinnamyl-3,4-dihydroxy-alpha-cyanocinnamate (10 microM) also inhibited the expression of c-fos and c-myc mRNA and, to a lesser extent, c-jun mRNA. CINNAMYL-3,4-DIHYDROXY-ALPHA-CYANOCINNAMATE 0-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 7621502-11 1994 A second group of rats were injected in opposing striata with biotin-labeled c-fos AS DNA and c-fos S DNA. Biotin 62-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 7621504-6 1994 Bilateral amygdala injection of a 15-mer phosphorothioate c-fos antisense oligodeoxynucleotide prior to testing blocked conflict-induced c-fos expression and had behavioral effects similar to those of established antianxiety drugs. Parathion 41-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 7621504-6 1994 Bilateral amygdala injection of a 15-mer phosphorothioate c-fos antisense oligodeoxynucleotide prior to testing blocked conflict-induced c-fos expression and had behavioral effects similar to those of established antianxiety drugs. Parathion 41-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 7621504-6 1994 Bilateral amygdala injection of a 15-mer phosphorothioate c-fos antisense oligodeoxynucleotide prior to testing blocked conflict-induced c-fos expression and had behavioral effects similar to those of established antianxiety drugs. Oligodeoxyribonucleotides 74-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 7621504-6 1994 Bilateral amygdala injection of a 15-mer phosphorothioate c-fos antisense oligodeoxynucleotide prior to testing blocked conflict-induced c-fos expression and had behavioral effects similar to those of established antianxiety drugs. Oligodeoxyribonucleotides 74-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 7965747-0 1994 Effects of chronic haloperidol and clozapine treatment on neurotensin and c-fos mRNA in rat neostriatal subregions. Haloperidol 19-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 7965747-0 1994 Effects of chronic haloperidol and clozapine treatment on neurotensin and c-fos mRNA in rat neostriatal subregions. Clozapine 35-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 7820657-4 1994 Allowing rats to drink water after AII infusions suppressed c-fos expression both AVP- and OXY-stained magnocellular neurons. Water 23-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 7820676-1 1994 Immunoreactivity against c-fos-like protein (FOS) was induced in many neuronal cell bodies within the sacral parasympathetic nucleus (SPN) in urethane-anesthetized rats by chemical irritation of the urinary bladder with formalin. Urethane 142-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-43 7820676-1 1994 Immunoreactivity against c-fos-like protein (FOS) was induced in many neuronal cell bodies within the sacral parasympathetic nucleus (SPN) in urethane-anesthetized rats by chemical irritation of the urinary bladder with formalin. Urethane 142-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 7965747-5 1994 An acute challenge of haloperidol 24 hr after chronic haloperidol treatment did not affect the tolerant response of NT neurons but caused a small increase in c-fos mRNA in the DLSt. Haloperidol 22-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 7820676-1 1994 Immunoreactivity against c-fos-like protein (FOS) was induced in many neuronal cell bodies within the sacral parasympathetic nucleus (SPN) in urethane-anesthetized rats by chemical irritation of the urinary bladder with formalin. Formaldehyde 220-228 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-43 7820676-1 1994 Immunoreactivity against c-fos-like protein (FOS) was induced in many neuronal cell bodies within the sacral parasympathetic nucleus (SPN) in urethane-anesthetized rats by chemical irritation of the urinary bladder with formalin. Formaldehyde 220-228 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 7891846-0 1994 MK-801 prevents cyanide-induced changes of Fos levels in rat brain. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 7820367-3 1994 We explored two possible mechanisms responsible for this phenomenon: Corticosteroid feedback and alcohol-induced inhibition of hypothalamic neuronal activation (measured through changes in c-fos and NGFI-B mRNA levels). Alcohols 97-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 189-194 7891846-0 1994 MK-801 prevents cyanide-induced changes of Fos levels in rat brain. Cyanides 16-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 7891846-1 1994 The effect of acute cyanide intoxication on levels of transcriptional regulatory proteins Fos and c-Jun in rat cortex, hippocampus, cerebellum and brain stem was studied. Cyanides 20-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 7891846-2 1994 Western blot analysis showed a differential effect of cyanide on Fos levels in the selected brain areas. Cyanides 54-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 7891846-10 1994 Pretreatment with the NMDA receptor antagonist, MK-801, prevented the cyanide-induced changes of Fos. Dizocilpine Maleate 48-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 7891846-10 1994 Pretreatment with the NMDA receptor antagonist, MK-801, prevented the cyanide-induced changes of Fos. Cyanides 70-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 7891846-11 1994 The differential effect of cyanide on Fos levels in different brain areas and the blockade of these changes by MK-801 suggest involvement of multiple neuronal pathways, including the excitatory amino acid (EAA) neurotransmitter system. Cyanides 27-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 7891846-12 1994 It is concluded that cyanide alters levels of the transcriptional regulatory protein Fos through activation of the EAA neurotransmitter system and, thus, may affect gene expression in neuronal or glia cells. Cyanides 21-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 7969045-2 1994 One early cellular response to cocaine administration is a brain region-specific alteration in the transcriptional pattern of immediate early genes belonging to the Fos/Jun family of nucleotide sequence-specific [activator protein-1 (AP-1)] DNA-binding proteins. Cocaine 31-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-168 7965748-7 1994 In contrast, measurements of c-fos, a protooncogene that couples cyclic AMP to induction of ODC, showed increased responses to beta adrenergic or cyclic AMP stimulation in PTU-treated animals. Cyclic AMP 65-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 7965748-7 1994 In contrast, measurements of c-fos, a protooncogene that couples cyclic AMP to induction of ODC, showed increased responses to beta adrenergic or cyclic AMP stimulation in PTU-treated animals. Cyclic AMP 146-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 7965748-7 1994 In contrast, measurements of c-fos, a protooncogene that couples cyclic AMP to induction of ODC, showed increased responses to beta adrenergic or cyclic AMP stimulation in PTU-treated animals. Propylthiouracil 172-175 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 7965748-8 1994 The effect of PTU on c-fos responsiveness occurred in the absence of alterations in basal c-fos expression, a situation different from that seen with adenylate cyclase or ODC. Propylthiouracil 14-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 7839314-0 1994 Acute administration of alcohol blocks cocaine-induced striatal c-fos immunoreactivity protein in the rat. Alcohols 24-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 7839314-0 1994 Acute administration of alcohol blocks cocaine-induced striatal c-fos immunoreactivity protein in the rat. Cocaine 39-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 7839314-1 1994 Immediate-early genes, such as c-fos, are induced in the brain by cocaine and other psychotropic drugs. Cocaine 66-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 7839314-3 1994 Because alcohol selectively blocks NMDA receptor function, we determined the ability of alcohol to block the expression of c-fos normally induced by systemic cocaine exposure in perikarya of the rat striatum. Alcohols 8-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 7839314-3 1994 Because alcohol selectively blocks NMDA receptor function, we determined the ability of alcohol to block the expression of c-fos normally induced by systemic cocaine exposure in perikarya of the rat striatum. Alcohols 88-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 7839314-3 1994 Because alcohol selectively blocks NMDA receptor function, we determined the ability of alcohol to block the expression of c-fos normally induced by systemic cocaine exposure in perikarya of the rat striatum. Cocaine 158-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 7839314-5 1994 Separate administration of three doses of alcohol alone (1, 2, or 3 g/kg) was ineffectual in inducing FOS-like protein in this or other regions of the rat brain. Alcohols 42-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 7839314-7 1994 Furthermore, the blockade of cocaine-induced FOS-like protein by alcohol occurred at a dose which produced a blood alcohol concentration of approximately 180 mg/dl (40 mM), comparable to that detected in intoxicating humans. Cocaine 29-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 7839314-7 1994 Furthermore, the blockade of cocaine-induced FOS-like protein by alcohol occurred at a dose which produced a blood alcohol concentration of approximately 180 mg/dl (40 mM), comparable to that detected in intoxicating humans. Alcohols 65-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 7839314-7 1994 Furthermore, the blockade of cocaine-induced FOS-like protein by alcohol occurred at a dose which produced a blood alcohol concentration of approximately 180 mg/dl (40 mM), comparable to that detected in intoxicating humans. Alcohols 115-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 7969045-4 1994 In the striatum, acute cocaine administration increases cellular levels of c-fos and jun-B mRNA, whereas transcriptional effects in the cerebellum are limited to c-fos mRNA. Cocaine 23-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 7969045-5 1994 After chronic cocaine treatment a desensitization of c-fos mRNA induction is observed in the striatum, with sensitization of the same transcriptional effect occurring in the cerebellum. Cocaine 14-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 7969045-7 1994 Gel retention analysis using antibodies to the various Fos and Jun proteins was used to characterize cocaine-dependent alterations in the composition of striatal and cerebellar AP-1 DNA-binding complexes. Cocaine 101-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 7969045-8 1994 In striatum, cocaine increases the relative levels of c-Fos, Fos-B, Jun-B, and Jun-D proteins that bind the AP-1 DNA sequence element, whereas in the cerebellum only c-Fos and Jun-D binding activities are increased. Cocaine 13-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 7937819-3 1994 Increased striatal c-fos expression ipsilateral to a unilateral lesion of the substantia nigra in rats treated with appropriate dopamine agonists provides a cellular marker of D1 receptor supersensitivity. Dopamine 128-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 7845598-8 1994 In contrast, parallel experiments showed that c-Fos does form a major component of the hippocampal activator protein-1 complex that is induced in rats following kainic acid treatment. Kainic Acid 161-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 7937819-5 1994 Because expression of the c-fos gene in response to cAMP- and Ca2+/calmodulin-regulated protein kinases depends on phosphorylation of cAMP-response element-binding protein (CREB) at Ser-133, we examined CREB phosphorylation after dopaminergic stimulation in cultured striatal neurons and in the striatum of rats after unilateral 6-hydroxydopamine ablation of the substantia nigra. Cyclic AMP 52-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 7937819-5 1994 Because expression of the c-fos gene in response to cAMP- and Ca2+/calmodulin-regulated protein kinases depends on phosphorylation of cAMP-response element-binding protein (CREB) at Ser-133, we examined CREB phosphorylation after dopaminergic stimulation in cultured striatal neurons and in the striatum of rats after unilateral 6-hydroxydopamine ablation of the substantia nigra. Serine 182-185 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 7937819-5 1994 Because expression of the c-fos gene in response to cAMP- and Ca2+/calmodulin-regulated protein kinases depends on phosphorylation of cAMP-response element-binding protein (CREB) at Ser-133, we examined CREB phosphorylation after dopaminergic stimulation in cultured striatal neurons and in the striatum of rats after unilateral 6-hydroxydopamine ablation of the substantia nigra. Oxidopamine 329-346 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 8092325-0 1994 Comparison of fos-like immunoreactivity induced in rat brain by central injection of angiotensin II and carbachol. Carbachol 104-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 7820626-8 1994 Compared to saline infused rats, phenylephrine infusion induced a significant increase in the proportion of those neurons that expressed Fos (14% vs. 1% P < 0.000.1). Phenylephrine 33-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-140 7824204-2 1994 Noxious stimulation of the hindpaw with formalin induces c-Fos in neurons in superficial laminae (I and II) of these dorsal horn segments. Formaldehyde 40-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 7808231-1 1994 During lactation, there is an inhibition of cortical and hippocampal activation in response to N-methyl-D,L-aspartate (NMA), but not kainate, as assessed by induction of c-Fos expression. n-methyl-d,l-aspartate 95-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-175 7808231-1 1994 During lactation, there is an inhibition of cortical and hippocampal activation in response to N-methyl-D,L-aspartate (NMA), but not kainate, as assessed by induction of c-Fos expression. nma 119-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-175 7808231-8 1994 During lactation, NMA induced c-Fos expression in similar areas of the brainstem as during the cycle, except in the locus coeruleus and dorsal raphe, where c-Fos expression was significantly less than that observed during the cycle. nma 18-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 7808231-8 1994 During lactation, NMA induced c-Fos expression in similar areas of the brainstem as during the cycle, except in the locus coeruleus and dorsal raphe, where c-Fos expression was significantly less than that observed during the cycle. nma 18-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 8063876-1 1994 Middle cerebral artery (MCA) occlusion in halothane-anesthetized rats induced c-fos, junB, and c-jun immediate early gene mRNAs and hsp70 heat shock gene mRNA in brain. Halothane 42-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 8087951-15 1994 Despite equivalent elevation of LV systolic pressure, fosinopril resulted in regression of myocyte hypertrophy in Fos/LVH versus LVH (myocyte cell width, 14.8 +/- 0.5 versus 20.8 +/- 2.2 microns, P < .05) to normal levels (Sham, 16.3 +/- 0.9 microns). Fosinopril 54-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 8082733-5 1994 The results show that increasing intracellular levels of cAMP by exposing the cells to the synthetic adenosine analogue N-ethyl-carboxamido adenosine (NECA) results in an accumulation of c-jun and c-fos mRNA in both cell types. Cyclic AMP 57-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 197-202 8082733-5 1994 The results show that increasing intracellular levels of cAMP by exposing the cells to the synthetic adenosine analogue N-ethyl-carboxamido adenosine (NECA) results in an accumulation of c-jun and c-fos mRNA in both cell types. Adenosine 101-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 197-202 8082733-5 1994 The results show that increasing intracellular levels of cAMP by exposing the cells to the synthetic adenosine analogue N-ethyl-carboxamido adenosine (NECA) results in an accumulation of c-jun and c-fos mRNA in both cell types. Adenosine-5'-(N-ethylcarboxamide) 120-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 197-202 8082733-5 1994 The results show that increasing intracellular levels of cAMP by exposing the cells to the synthetic adenosine analogue N-ethyl-carboxamido adenosine (NECA) results in an accumulation of c-jun and c-fos mRNA in both cell types. Adenosine-5'-(N-ethylcarboxamide) 151-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 197-202 7916044-1 1994 Prenatal treatment with the D1-dopamine receptor agonist SKF 38393 or cocaine induces expression of the immediate-early gene c-fos in the fetal rat suprachiasmatic nucleus (SCN) (Weaver et al., 1992). 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 57-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 7916044-1 1994 Prenatal treatment with the D1-dopamine receptor agonist SKF 38393 or cocaine induces expression of the immediate-early gene c-fos in the fetal rat suprachiasmatic nucleus (SCN) (Weaver et al., 1992). Cocaine 70-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 7830888-7 1994 In the neostriatum, eticlopride caused a rapid increase in c-fos messenger RNA with significantly increased levels at 10 min (P < 0.01). eticlopride 20-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 8083758-3 1994 It has recently been demonstrated that amphetamine regulates the expression of several genes, including c-fos, via dopamine D1 receptors in rat striatum. Amphetamine 39-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 8083758-5 1994 In addition, we show by antisense injection that CREB is necessary for c-fos induction by amphetamine in vivo. Amphetamine 90-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 7825122-6 1994 Image analysis of film autoradiograms demonstrated that 1 h after a single injection of cocaine, the expression of c-fos and zif/268, but not PPD or PPE in the dorsal striatum and cortex, was enhanced in a dose-dependent manner as compared to that in saline controls. Cocaine 88-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 7825122-6 1994 Image analysis of film autoradiograms demonstrated that 1 h after a single injection of cocaine, the expression of c-fos and zif/268, but not PPD or PPE in the dorsal striatum and cortex, was enhanced in a dose-dependent manner as compared to that in saline controls. Sodium Chloride 251-257 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 8073469-7 1994 Pretreatment with MK-801 (3 mg/kg) largely inhibited the induction of c-fos mRNA, indicating that the induction was mediated through an N-methyl-D-aspartate subtype of glutamate receptor. Dizocilpine Maleate 18-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 7717068-4 1994 The results indicated that 0.1 mumol.L-1 sincalide induced c-fos expression markedly in both brain (a 3.8-fold increase in c-fos protein level) and in spinal dorsal horn (a 3.6-fold increase). Sincalide 41-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 7717068-4 1994 The results indicated that 0.1 mumol.L-1 sincalide induced c-fos expression markedly in both brain (a 3.8-fold increase in c-fos protein level) and in spinal dorsal horn (a 3.6-fold increase). Sincalide 41-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 7717068-5 1994 NDAP (a kappa receptor agonist) 0.1 mumol.L-1 showed some activating effects on c-fos expression, the c-fos protein level increased 2.7 and 2.6 times respectively in brain and spinal dorsal horn. ndap 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 7717068-5 1994 NDAP (a kappa receptor agonist) 0.1 mumol.L-1 showed some activating effects on c-fos expression, the c-fos protein level increased 2.7 and 2.6 times respectively in brain and spinal dorsal horn. ndap 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 7717068-6 1994 Ohmefentanyl (Ohm, a mu receptor agonist) 0.1 mumol.L-1 also exhibited an inducing effect on c-fos protein production. F 7302 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 7717068-8 1994 In contrast, the effect of sincalide on c-fos protein production is antagonistic to that of Ohm. Sincalide 27-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 7816320-2 1994 The expression of c-fos in the PVN appeared about 3 h after insulin treatment and was very high after 5 h while no labelling was observed in isotonic saline-injected animals. Sodium Chloride 150-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 7520433-5 1994 Preincubation with the calcineurin inhibitors FK-506 or cyclosporin A strongly enhanced expression of NGFI-A and blocked transcription of NGFI-B, but it had no significant effect on Ca(2+)-stimulated transcription of c-fos. Tacrolimus 46-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 217-222 7520433-5 1994 Preincubation with the calcineurin inhibitors FK-506 or cyclosporin A strongly enhanced expression of NGFI-A and blocked transcription of NGFI-B, but it had no significant effect on Ca(2+)-stimulated transcription of c-fos. Cyclosporine 56-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 217-222 7983241-5 1994 The reduction in Fos-like immunoreactivity by vagal stimulation was abolished by intrathecal administration of methysergide, a nonselective serotoninergic receptor antagonist, but not by phentolamine, a nonselective alpha-adrenoceptor antagonist. Methysergide 111-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 7982096-0 1994 Action of angiotensin converting enzyme inhibitors in rat brain: interaction with isoproterenol assessed by Fos immunocytochemistry. Isoproterenol 82-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 8060343-3 1994 We find that genistein treatment prevents phenylephrine-induced activation of three promoters (Fos, atrial natriuretic factor, ANF, and the myosin light chain 2, MLC-2), which are activated in the hypertrophic response. Genistein 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 8060343-3 1994 We find that genistein treatment prevents phenylephrine-induced activation of three promoters (Fos, atrial natriuretic factor, ANF, and the myosin light chain 2, MLC-2), which are activated in the hypertrophic response. Phenylephrine 42-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 8062219-0 1994 Induction of c-fos and c-myc oncogene expression in the pyloric mucosa of rat stomach by N-methyl-N"-nitro-N-nitrosoguanidine and taurocholate. Methylnitronitrosoguanidine 89-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 8062219-0 1994 Induction of c-fos and c-myc oncogene expression in the pyloric mucosa of rat stomach by N-methyl-N"-nitro-N-nitrosoguanidine and taurocholate. Taurocholic Acid 130-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 8080455-2 1994 The serine/threonine protein phosphatase inhibitor okadaic acid (OA) was found to enhance mRNA transcripts of c-fos and of the jun family of proto-oncogenes including c-jun, jun B and jun D in cultured pheochromocytoma PC12 cells. Okadaic Acid 51-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 8080455-2 1994 The serine/threonine protein phosphatase inhibitor okadaic acid (OA) was found to enhance mRNA transcripts of c-fos and of the jun family of proto-oncogenes including c-jun, jun B and jun D in cultured pheochromocytoma PC12 cells. Okadaic Acid 65-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 7819539-5 1994 administered NMDA exhibit differential specificity for anatomical regions of the brain in stimulating c-fos. N-Methylaspartate 13-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 7819539-8 1994 NMDA produced behaviorally similar clonic (popcorn) convulsions associated with transient increases in c-fos mRNA in different brain areas. N-Methylaspartate 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 7819539-10 1994 However, the route of administration clearly influenced the anatomical specificity of the NMDA-induced c-fos mRNA changes. N-Methylaspartate 90-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 7819539-14 1994 NMDA produced maximal c-fos mRNA stimulation in the cerebral cortex. N-Methylaspartate 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 7990028-13 1994 The level of c-fos mRNA also increased transiently during repeated KCl-induced depolarizations but c-jun mRNA remained low or absent. Potassium Chloride 67-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 7982066-4 1994 In water-replete rats, Fos-IR was absent or very low in these regions. Water 3-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 7915496-4 1994 The products of some of these genes, such as c-fos and c-jun, are known to form a heterodimeric transcription factor complex (AP-1) that binds specifically to the consensus sequence TGAC(G)TCA. Trichloroacetic Acid 189-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 7915496-6 1994 In both parietal and GH3 cells, c-fos-specific mRNA was increased by agents known to act via both adenosine-3",5"-cyclic monophosphate and Ca(2+)-dependent signaling mechanisms, and octreotide significantly inhibited this response. Cyclic AMP 98-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 7915496-6 1994 In both parietal and GH3 cells, c-fos-specific mRNA was increased by agents known to act via both adenosine-3",5"-cyclic monophosphate and Ca(2+)-dependent signaling mechanisms, and octreotide significantly inhibited this response. Octreotide 182-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 7953748-0 1994 Induction of Fos protein in the piriform cortex after brain injury in pentobarbital-anaesthetized rats: lack of effect of lactation. Pentobarbital 70-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 7825122-1 1994 The effects of short- and long-term cocaine exposure on the expression of the nuclear transcription factor genes, c-fos and zif/268, as well as the opioid peptides, preprodynorphin (PPD) and preproenkephalin (PPE), in various regions of rat brain were evaluated by injecting i.p. Cocaine 36-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 7953732-9 1994 Fos was induced in both SON and PVN of all water deprived rats regardless of age. Water 43-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 7835407-9 1994 H7 (50 microM), genistein (2 microM) and okadaic acid (20 nM), all block the induction of c-jun and c-fos mRNA accumulation in the H2O2-treated rat lenses. Genistein 16-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 7835407-9 1994 H7 (50 microM), genistein (2 microM) and okadaic acid (20 nM), all block the induction of c-jun and c-fos mRNA accumulation in the H2O2-treated rat lenses. Okadaic Acid 41-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 7835407-9 1994 H7 (50 microM), genistein (2 microM) and okadaic acid (20 nM), all block the induction of c-jun and c-fos mRNA accumulation in the H2O2-treated rat lenses. Hydrogen Peroxide 131-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 7835407-10 1994 These results suggest that H2O2 activates protein kinase and phosphatase dependent signal transduction pathways to induce c-jun and c-fos expression which may regulate lens crystallin genes and other genes containing AP-1 binding sites. Hydrogen Peroxide 27-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 7845612-6 1994 Reserpine rendered striatal D1 and D2 receptors supersensitive as indicated by 10- to 12-fold increases in striatal and pallidal Fos immunoreactivity. Reserpine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-132 7984056-4 1994 Treatment with pilocarpine (30 mg/kg) alone increased mRNA levels in the order of c-fos > jun-B > c-jun but did not change the jun-D mRNA level, and maximal c-fos and jun-B mRNA levels occurred earlier (30 min) in the cortex than in the hippocampus. Pilocarpine 15-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 7984056-4 1994 Treatment with pilocarpine (30 mg/kg) alone increased mRNA levels in the order of c-fos > jun-B > c-jun but did not change the jun-D mRNA level, and maximal c-fos and jun-B mRNA levels occurred earlier (30 min) in the cortex than in the hippocampus. Pilocarpine 15-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-168 7984056-5 1994 Treatment with the lower dose of pilocarpine (5 mg/kg) alone caused only small increases in c-fos and jun-B mRNA levels and these responses were unaffected by lithium pretreatment. Pilocarpine 33-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 7984056-7 1994 The mRNA levels were increased during seizures in the order of c-fos > jun-B > c-jun > jun-D in the hippocampus and jun-B > c-fos > c-jun > jun-D in the cortex, and were increased for a longer duration as well as to a greater extent than after administration of pilocarpine alone. Pilocarpine 280-291 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 7984056-7 1994 The mRNA levels were increased during seizures in the order of c-fos > jun-B > c-jun > jun-D in the hippocampus and jun-B > c-fos > c-jun > jun-D in the cortex, and were increased for a longer duration as well as to a greater extent than after administration of pilocarpine alone. Pilocarpine 280-291 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 7953748-1 1994 The ability of lactation and progesterone administration to inhibit the induction of Fos protein in the piriform cortex after brain injury in pentobarbital-anaesthetized rats was assessed in three studies. Pentobarbital 142-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 7953748-2 1994 Consistent with previous reports we found that brain injury-induced Fos expression in the piriform cortex could be eliminated by the administration of the non-competitive NMDA receptor antagonist MK 801 (4 mg/kg i.p.). Dizocilpine Maleate 196-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 8035171-6 1994 An analysis of the effects of IL-1 and TPA on immediate early gene expression indicated that IL-1 preferentially induced c-jun gene expression, whereas TPA greatly increased c-fos and zif/268 gene expression. Tetradecanoylphorbol Acetate 152-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 174-179 7804603-0 1994 c-fos antisense oligonucleotide specifically attenuates haloperidol-induced increases in neurotensin/neuromedin N mRNA expression in rat dorsal striatum. Oligonucleotides 16-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 7804603-0 1994 c-fos antisense oligonucleotide specifically attenuates haloperidol-induced increases in neurotensin/neuromedin N mRNA expression in rat dorsal striatum. Haloperidol 56-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 7804603-3 1994 Our recent studies demonstrate that a vast majority of haloperidol-sensitive NT/N mRNA expressing cells in the dorsolateral striatum coexpress c-fos mRNA. Haloperidol 55-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-148 7804603-9 1994 These data demonstrate a specific role of Fos in the regulation of the neurotensin/neuromedin N gene in the rat dorsolateral striatal neurons by acute haloperidol treatment. Haloperidol 151-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 7994657-1 1994 We use NMDA to induce expression of c-fos mRNA as a marker to observe the activity of NMDA receptor in neurons during development, and compare the activity of NMDA receptor between audiogenic epilepsy -prone (P77PMC) and audiogenic epilepsy resistant rats brain. N-Methylaspartate 7-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 7994657-2 1994 In primary culture of rats cerebral cortical neurons NMDA induced c-fos mRNA expression exhibits dose and time-dependent changes, which can be prevented by antagonists. N-Methylaspartate 53-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 7994657-3 1994 During the development of neurons, the NMDA -induced c-fos mRNA expression reaches a maximum at day 24. N-Methylaspartate 39-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 7994657-4 1994 NMDA-induced c-fos mRNA expression of P77PMC rats is higher than that of controls during 6 to 24 days in vitro with significant difference (P < 0.05) at day 18. N-Methylaspartate 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 7994657-5 1994 To present changes in c-fos mRNA expression induced by NMDA in cultured P77PMC rat cortical neurons may be one of the factors related to susceptibility of epilepsy in P77PMC rats. N-Methylaspartate 55-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 7953725-4 1994 In adult rats whose striatum had been lesioned with ibotenic acid 10-12 days prior to implantation of fetal striatal tissue, 3- and 18-month-old striatal grafts showed Fos immunoreactivity in a considerable number of cells after either bilateral, or ipsilateral (approximately 30-40% of the density of Fos-immunoreactive cells in the normal striatum) or contralateral cortical stimulation. Ibotenic Acid 52-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-171 7953725-4 1994 In adult rats whose striatum had been lesioned with ibotenic acid 10-12 days prior to implantation of fetal striatal tissue, 3- and 18-month-old striatal grafts showed Fos immunoreactivity in a considerable number of cells after either bilateral, or ipsilateral (approximately 30-40% of the density of Fos-immunoreactive cells in the normal striatum) or contralateral cortical stimulation. Ibotenic Acid 52-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 302-305 7922580-1 1994 We have previously demonstrated that Fos immunoreactivity can be stimulated by KCl, forskolin or glutamate in cultured tyrosine hydroxylase-immunoreactive (TH-ir) hypothalamic neurons. Potassium Chloride 79-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 7922580-1 1994 We have previously demonstrated that Fos immunoreactivity can be stimulated by KCl, forskolin or glutamate in cultured tyrosine hydroxylase-immunoreactive (TH-ir) hypothalamic neurons. Colforsin 84-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 7922580-1 1994 We have previously demonstrated that Fos immunoreactivity can be stimulated by KCl, forskolin or glutamate in cultured tyrosine hydroxylase-immunoreactive (TH-ir) hypothalamic neurons. Glutamic Acid 97-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 7922580-6 1994 Pretreatment of the cultures with the D2 agonist LY163502 inhibited KCl- and forskolin-stimulated Fos-ir in TH-ir neurons in a saturable dose-dependent manner. quinelorane 49-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 7922580-6 1994 Pretreatment of the cultures with the D2 agonist LY163502 inhibited KCl- and forskolin-stimulated Fos-ir in TH-ir neurons in a saturable dose-dependent manner. Potassium Chloride 68-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 7922580-6 1994 Pretreatment of the cultures with the D2 agonist LY163502 inhibited KCl- and forskolin-stimulated Fos-ir in TH-ir neurons in a saturable dose-dependent manner. Colforsin 77-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 7922580-7 1994 The maximal effective dose was 30 microM LY163502, which decreased Fos-ir by 23% in cultures treated with 50 mM KCl and by 33% in those treated with 30 microM forskolin. quinelorane 41-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 7922580-7 1994 The maximal effective dose was 30 microM LY163502, which decreased Fos-ir by 23% in cultures treated with 50 mM KCl and by 33% in those treated with 30 microM forskolin. Potassium Chloride 112-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 7922580-9 1994 LY163502 inhibition of Fos-ir was blocked by D2 antagonist or Bordetella pertussis toxin pretreatment which demonstrates that the effect is mediated by D2 receptor activation of an inhibitory G protein. quinelorane 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 7970238-4 1994 Urethane induced the largest amount of Fos-ir neurons (n = 8439), while fentanyl/midazolam the smallest (n = 112). Urethane 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 7970238-6 1994 The results indicate that fentanyl/midazolam is the most recommendable anesthetic drug in the Fos expression study. Fentanyl 26-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 7970238-6 1994 The results indicate that fentanyl/midazolam is the most recommendable anesthetic drug in the Fos expression study. Midazolam 35-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 8048943-2 1994 Elevation of cellular polyamine content triggered the transcription of c-myc and c-fos. Polyamines 22-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 7970213-1 1994 The immunoreactivity of c-fos protein was transiently detected in magnocellular neurons of the supraoptic nucleus (SON) and paraventricular nucleus (PVN) of the rat hypothalamus after intraperitoneal injection of hypertonic NaCl solution. nacl solution 224-237 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 8048943-3 1994 alpha-Difluoromethylornithine, a specific inhibitor of ODC, prevented the transcription of c-myc in cells grown at 37 degrees C. Putrescine, at physiological concentrations, triggered the transcription of c-myc and c-fos in cells grown at 42 degrees C, when Ki-ras was not expressed. Eflornithine 0-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 215-220 8048943-3 1994 alpha-Difluoromethylornithine, a specific inhibitor of ODC, prevented the transcription of c-myc in cells grown at 37 degrees C. Putrescine, at physiological concentrations, triggered the transcription of c-myc and c-fos in cells grown at 42 degrees C, when Ki-ras was not expressed. Putrescine 129-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 215-220 7948416-2 1994 It has been found that only the sublethal (3 mg/kg) dose of cycloheximide which induces the superexpression of c-fos and c-myc oncogenes can promote sphingosine accumulation in the cell. Cycloheximide 60-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 7948416-2 1994 It has been found that only the sublethal (3 mg/kg) dose of cycloheximide which induces the superexpression of c-fos and c-myc oncogenes can promote sphingosine accumulation in the cell. Sphingosine 149-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 8040263-3 1994 Phenylephrine, a selective alpha 1 adrenergic receptor agonist, caused a rapid and transient increase in c-fos mRNA accumulation which was inhibited by prazosin, an alpha 1 receptor antagonist. Phenylephrine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 8040263-3 1994 Phenylephrine, a selective alpha 1 adrenergic receptor agonist, caused a rapid and transient increase in c-fos mRNA accumulation which was inhibited by prazosin, an alpha 1 receptor antagonist. Prazosin 152-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 8040263-5 1994 Chloroethyl-clonidine, a compound which irreversibly blocks alpha 1B receptors, completely blocked the phenylephrine-induced increase in c-fos mRNA. chlorethylclonidine 0-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 8040263-5 1994 Chloroethyl-clonidine, a compound which irreversibly blocks alpha 1B receptors, completely blocked the phenylephrine-induced increase in c-fos mRNA. Phenylephrine 103-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 8040263-7 1994 Phenylephrine-induced c-fos mRNA was partially inhibited by H-7, a protein kinase C inhibitor, and by nifedipine, a Ca2+ channel blocker; these two compounds together had additive effects. Phenylephrine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 8040263-7 1994 Phenylephrine-induced c-fos mRNA was partially inhibited by H-7, a protein kinase C inhibitor, and by nifedipine, a Ca2+ channel blocker; these two compounds together had additive effects. 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine 60-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 8040263-7 1994 Phenylephrine-induced c-fos mRNA was partially inhibited by H-7, a protein kinase C inhibitor, and by nifedipine, a Ca2+ channel blocker; these two compounds together had additive effects. Nifedipine 102-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 8040263-8 1994 In situ hybridization showed that expression of c-fos mRNA induced by phenylephrine was localized to aorta"s medial layer. Phenylephrine 70-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 7953655-0 1994 Formalin induced FOS-like immunoreactive neurons in the trigeminal spinal caudal subnucleus project to contralateral parabrachial nucleus in the rat. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 7953655-5 1994 The results suggest that FOS-like immunoreactivity might serve as an indicator for the nociceptive response after formalin injection into the trigeminal region and that the PB might be an important relay station for the further processing of the nociceptive information relayed from the trigeminal afferents. Formaldehyde 114-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 8088356-6 1994 The number of Fos-like immunoreactive neurons induced by the heat stimulation was significantly decreased by pretreatment with morphine (42, 64 and 75% decrease as compared to control values after 2.5, 5 and 7.5 mg/kg i.v. Morphine 127-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 8088356-12 1994 RB 101, a systemically active mixed inhibitor of enkephalin-metabolising enzymes, significantly decreased Fos-like immunoreactivity induced by heat stimulation (19, 29 and 48% decreases as compared to control values at 10, 20 and 40 mg/kg i.v. RB 101 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 8206384-3 1994 The nucleotide (nt) sequence of the 5"-flanking region of the rat c-fos gene displays remarkable similarity with the mouse and human c-fos genes (93 and 77% identity, respectively). Nucleotides 16-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 8077463-5 1994 Injection of apomorphine (5 mg/kg) resulted in increased expression of both c-fos and zif268 immediate-early genes in cortex and striatum. Apomorphine 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 7919167-0 1994 Forskolin increases phosphorylated-CREB and fos immunoreactivity in rat striatum. Colforsin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 7919167-4 1994 Although forskolin did not alter CREB-IR, forskolin did induce striatal P-CREB-IR and Fos-IR by 2.5- and 10-fold, respectively. Colforsin 42-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 7919180-5 1994 Twenty minutes after a single THC injection, significant increases in concentration of the mRNAs for C-FOS, C-JUN and ZIF-268 were observed in the cingulate cortex (75, 45 and 37%) and for C-FOS and ZIF-268 in the fronto-parietal cortex (60 and 64%) and caudate-putamen (81 and 32%) while JUN-D mRNA levels were not changed. Dronabinol 30-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 7919180-5 1994 Twenty minutes after a single THC injection, significant increases in concentration of the mRNAs for C-FOS, C-JUN and ZIF-268 were observed in the cingulate cortex (75, 45 and 37%) and for C-FOS and ZIF-268 in the fronto-parietal cortex (60 and 64%) and caudate-putamen (81 and 32%) while JUN-D mRNA levels were not changed. Dronabinol 30-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 189-194 7913445-7 1994 Amphetamine produced widespread induction of the immediate-early gene c-fos in cells of host striatum that extended beyond the transplant-derived DA innervation. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 7835407-0 1994 The redox active components H2O2 and N-acetyl-L-cysteine regulate expression of c-jun and c-fos in lens systems. Hydrogen Peroxide 28-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 7835407-0 1994 The redox active components H2O2 and N-acetyl-L-cysteine regulate expression of c-jun and c-fos in lens systems. Acetylcysteine 37-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 7835407-4 1994 At optimal concentrations of H2O2, mRNA accumulation of c-jun and c-fos in the rat lenses is induced 20- and 18-fold above normal levels respectively, but with distinct kinetics. Hydrogen Peroxide 29-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 7835407-7 1994 The antioxidant N-acetyl-cysteine (NAC) has a dual effect on the induction of c-jun and c-fos. Acetylcysteine 16-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 7835407-7 1994 The antioxidant N-acetyl-cysteine (NAC) has a dual effect on the induction of c-jun and c-fos. Acetylcysteine 35-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 7835407-9 1994 H7 (50 microM), genistein (2 microM) and okadaic acid (20 nM), all block the induction of c-jun and c-fos mRNA accumulation in the H2O2-treated rat lenses. 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine 0-2 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 7968354-2 1994 Recently it has been demonstrated that the centrally active muscarinic receptor agonist pilocarpine induces both c-fos mRNA and protein in rat brain. Pilocarpine 88-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 7968354-4 1994 Pretreatment of animals with atropine or pirenzepine significantly reduced induction of c-fos, jun-B, krox-20 and krox-24 genes in both hippocampus and cortex. Atropine 29-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 7968354-4 1994 Pretreatment of animals with atropine or pirenzepine significantly reduced induction of c-fos, jun-B, krox-20 and krox-24 genes in both hippocampus and cortex. Pirenzepine 41-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 7968356-0 1994 A single administration of ethanol simultaneously increases c-fos mRNA and reduces c-jun mRNA in the hypothalamus and hippocampus. Ethanol 27-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 7968356-1 1994 We have previously demonstrated that a single administration of ethanol induces the expression of c-fos mRNA in the hypothalamic paraventricular nucleus (PVN). Ethanol 64-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 7968356-5 1994 Using in situ hybridization histochemistry with oligonucleotide probes, we found that ethanol increased c-fos mRNA in the PVN, but decreased c-jun mRNA both in the PVN and in hippocampus. Ethanol 86-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 7968356-7 1994 Both cold and ethanol increased c-fos mRNA, and the effects were additive. Ethanol 14-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 7968357-0 1994 Induction of c-fos and CRF mRNA by MK-801 in the parvocellular paraventricular nucleus of the rat hypothalamus. Dizocilpine Maleate 35-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 7968357-4 1994 MK-801 markedly increased the expression of Fos-like protein in parvocellular nerve cells of the PVN within 60 min of systemic treatment, and double labeling immunocytochemistry indicated that Fos was primarily localized in CRF-containing neurons of the PVN. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 7968357-5 1994 MK-801 also significantly increased CRF biosynthesis as detected by in situ hybridization, thus suggesting that c-fos could be involved in the regulation of CRF genes. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 7968367-3 1994 Kainate (12 mg/kg) administration induced a biphasic activation of AP-1 in rat cerebral cortex and hippocampus with maximal levels observed at 1.5 h and 4.5 h and lower levels at 3 h and 6 h. Kainate also induced biphasic increases in the concentrations of some of the AP-1 constituent proteins (immediate early gene protein products), with initial increases of c-Jun, Fos, and Jun B occurring at 1.5 h and secondary larger increases at 4.5 h, but the level of Jun D was not altered by kainate treatment. Kainic Acid 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 369-372 7968367-7 1994 Binding of the activated glucocorticoid receptor to c-Jun or Fos is likely to contribute to the decreased AP-1 DNA binding activity following dexamethasone treatment. Dexamethasone 142-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 8013086-2 1994 Pretreatment with TGF-beta 1 potentiated norepinephrine (NE)-induced and stretch-induced (+10% and +20% elongation, for 30 minutes) c-fos mRNA expression by 2.2-fold, whereas TGF-beta 1 alone did not induce c-fos mRNA expression in Northern blot analysis. Norepinephrine 41-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 8013086-5 1994 The protein kinase C activators (12-O-tetradecanoylphorbol 13-acetate [TPA], phorbol 12,13-dibutyrate, and 1-oleyl-2-acetyl-rac-glycerol) induced c-fos mRNA expression, which was also potentiated by TGF-beta 1. Tetradecanoylphorbol Acetate 33-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-151 8013086-5 1994 The protein kinase C activators (12-O-tetradecanoylphorbol 13-acetate [TPA], phorbol 12,13-dibutyrate, and 1-oleyl-2-acetyl-rac-glycerol) induced c-fos mRNA expression, which was also potentiated by TGF-beta 1. Phorbol 12,13-Dibutyrate 77-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-151 8013086-5 1994 The protein kinase C activators (12-O-tetradecanoylphorbol 13-acetate [TPA], phorbol 12,13-dibutyrate, and 1-oleyl-2-acetyl-rac-glycerol) induced c-fos mRNA expression, which was also potentiated by TGF-beta 1. 1-oleyl-2-acetyl-rac-glycerol 107-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-151 7521477-4 1994 Induction of c-fos and Egr-1 mRNA by AII and ET was completely blocked by nisoldipine. Nisoldipine 74-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 7933827-0 1994 High glucose elevates c-fos and c-jun transcripts and proteins in mesangial cell cultures. Glucose 5-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 7933827-4 1994 Using quantitative reverse transcription (RT) PCR, we now report that mRNA levels for c-fos and c-jun were increased approximately twofold after treatment with high glucose. Glucose 165-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 7933827-8 1994 Immunofluorescence studies with polyclonal antibodies to c-fos and c-jun revealed that high glucose treatment for four hours increased nuclear staining intensity two- to threefold for both proteins. Glucose 92-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 7933827-11 1994 Thus, high glucose may effect gene expression of ECM proteins by elevating the transcription factors c-fos and c-jun which complex with one another to form activator protein 1 (AP-1). Glucose 11-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 8014873-5 1994 In the forebrain of 2-day-old rats, intracisternal administration of isoproterenol, a beta adrenergic agonist, produced > 20-fold stimulation of c-fos within 10 min of drug administration. Isoproterenol 69-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 7970213-2 1994 In contrast, c-fos-positive magnocellular neurons were persistently observed in the SON and PVN of the rats which were chronically stimulated by the drinking of hypertonic NaCl solution instead of water or by water deprivation. nacl solution 172-185 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 7970213-2 1994 In contrast, c-fos-positive magnocellular neurons were persistently observed in the SON and PVN of the rats which were chronically stimulated by the drinking of hypertonic NaCl solution instead of water or by water deprivation. Water 197-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 8189226-7 1994 However, in the case of H2O2-treated cells, the accumulation of c-fos or c-jun mRNAs was still pronounced 6 h after the end of the treatment and the levels of cell-secreted NGF appeared relatively reduced, when compared with those obtained after a shock with the X/XOD system. Hydrogen Peroxide 24-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 7970213-2 1994 In contrast, c-fos-positive magnocellular neurons were persistently observed in the SON and PVN of the rats which were chronically stimulated by the drinking of hypertonic NaCl solution instead of water or by water deprivation. Water 209-214 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 8189226-11 1994 Furthermore, treatment of cells with the protein synthesis inhibitor anisomycin had an effect similar to that of H2O2 because it caused an accumulation of c-fos, c-jun, and NGF transcripts after 6 h of treatment. Anisomycin 69-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-160 8069690-0 1994 Induction of c-fos in rat brain by acute cocaine and fenfluramine exposure: a comparison study. Cocaine 41-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 8069690-0 1994 Induction of c-fos in rat brain by acute cocaine and fenfluramine exposure: a comparison study. Fenfluramine 53-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 8069690-6 1994 In contrast, fenfluramine, but not cocaine, elicited c-fos mRNA and Fos-like protein in the neuroendocrine paraventricular nucleus (PVN) of the hypothalamus despite the fact that both drugs are known to be equally capable to stimulate the hypothalamic-pituitary-adrenal (HPA) axis. Fenfluramine 13-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 8069690-8 1994 To further identify the phenotypes of nerve cells expressing c-fos by fenfluramine in the PVN, it was demonstrated that the immediate-early gene was induced in a subpopulation of neurons constitutively expressing nitric oxide synthase (NOS). Fenfluramine 70-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 8069690-9 1994 Taken together, we identified a number of common and disparate actions of cocaine and fenfluramine in striatal and hypothalamic tissue, thereby suggesting that c-fos induction in these two brain structures is differentially regulated by intrinsic events in addition to neuronal phenotype. Cocaine 74-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 8069690-9 1994 Taken together, we identified a number of common and disparate actions of cocaine and fenfluramine in striatal and hypothalamic tissue, thereby suggesting that c-fos induction in these two brain structures is differentially regulated by intrinsic events in addition to neuronal phenotype. Fenfluramine 86-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 8069673-2 1994 Previously, we found that vaginal-cervical stimulation (VCS), by mating or manual probing, increases the expression of Fos-immunoreactivity (Fos-IR) in discrete populations of neurons in the preoptic area, mediobasal hypothalamus and midbrain, suggesting that these neurons respond to VCS. VCP 56-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-122 8069673-2 1994 Previously, we found that vaginal-cervical stimulation (VCS), by mating or manual probing, increases the expression of Fos-immunoreactivity (Fos-IR) in discrete populations of neurons in the preoptic area, mediobasal hypothalamus and midbrain, suggesting that these neurons respond to VCS. VCP 56-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 8069673-4 1994 Contrary to our hypothesis, in Experiment 1 priming animals with a behaviorally effective dose of 17 beta-estradiol benzoate followed 48 h later by progesterone caused a trend towards a decrease in the number of VCS-induced Fos-IR cells in the ventromedial hypothalamus. estradiol-17 beta-benzoate 98-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 224-227 8069673-4 1994 Contrary to our hypothesis, in Experiment 1 priming animals with a behaviorally effective dose of 17 beta-estradiol benzoate followed 48 h later by progesterone caused a trend towards a decrease in the number of VCS-induced Fos-IR cells in the ventromedial hypothalamus. Progesterone 148-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 224-227 8055340-1 1994 The GABA agonist muscimol, injected into the depressor area of the caudal ventrolateral medulla, increased blood pressure and increased the expression of the immediate early gene c-fos in the rostral ventral medulla (RVM) of the rat. gamma-Aminobutyric Acid 4-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-184 7936109-2 1994 A rapid and transient induction of c-fos, NGFI-A and NGFI-B (nerve growth factor-induced genes A and B) mRNA was demonstrated in the nucleus tractus solitarius (NTS), area postrema (AP), hypothalamic paraventricular (PVN) and supraoptic (SON) nuclei, and central nucleus of the amygdala, following peripheral administration of CCK-8S (1-100 micrograms/kg i.p.). 8-sulfocholecystokinin octapeptide 327-333 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 7945571-0 1994 Acetaldehyde induces c-fos and c-jun proto-oncogenes in fat-storing cell cultures through protein kinase C activation. Acetaldehyde 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 7945571-3 1994 We here evaluated whether acetaldehyde increases Col I and FN gene transcription through the induction of c-fos and c-jun proto-oncogenes and studied the possible role played by protein kinase C (PKC) and c-AMP. Acetaldehyde 26-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 7945571-5 1994 Acetaldehyde produced a rapid and transient induction of fos mRNA (undetectable at t = 0, peak at t = 45 and still evident at t = 90). Acetaldehyde 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 7945571-8 1994 These inhibitors of PKC activity blocked the stimulatory effect of acetaldehyde on fos and jun mRNA expression. Acetaldehyde 67-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 7945571-12 1994 We conclude that acetaldehyde increases procollagen I and fibronectin gene transcription in FSCs, possibly through c-fos and c-jun expression, and that PKC may play a regulatory role in this chain of events. Acetaldehyde 17-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 8203532-1 1994 The current study determines the hypothesis that expression of protooncogenes c-fos and c-myc is involved in the mechanism of polyamine-stimulated healing in gastric mucosal stress ulcers. Polyamines 126-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 8203532-4 1994 Exposure to stress led to appearance of c-fos mRNA and oncoprotein in the gastric oxyntic gland mucosa at 2 h and its disappearance by 4 h. Baseline expression of c-myc was enhanced significantly after 6 h of stress and remained elevated for 4 h. This change in the expression of c-fos and c-myc mRNA and oncoprotein preceded an increased rate of [3H]thymidine incorporation into mucosal DNA. Tritium 348-350 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 8203532-6 1994 DFMO also completely inhibited the expression of c-fos and significantly decreased c-myc mRNA and oncoprotein in the gastric mucosa of stressed rats. Eflornithine 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 7914658-0 1994 Role of a 35 kDa fos-related antigen (FRA) in the long-term induction of striatal dynorphin expression in the 6-hydroxydopamine lesioned rat. Oxidopamine 110-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 7914658-11 1994 Consistent with the notion that Fos and FRA proteins alter transcriptional activity by binding to AP-1 (or AP-1-like) DNA sequences in the promoter regions of target genes, we found that repeated APO treatment caused large increases in AP-1 binding activity in striata ipsilateral to 6-OHDA lesions. Oxidopamine 284-290 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 7914658-13 1994 While co-administration of the NMDA receptor antagonist, MK-801, inhibited APO-induced increases in DYN and Fos/FRA expression in the intact striatum, its only effect in the DA-denervated striatum was a partial (35%) inhibition of the APO-induced increase in DYN-ir concentrations. Dizocilpine Maleate 57-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 7914659-0 1994 Coexpression of neurotensin and c-fos mRNAs in rat neostriatal neurons following acute haloperidol. Haloperidol 87-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 7914659-3 1994 The present study used double-labeling in situ hybridization technique to study cellular localization of NT/N and c-fos mRNA following acute haloperidol treatment. Haloperidol 141-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 7914659-5 1994 Interestingly, approximately 75% of DLSt neurons expressing NT/N mRNA also displayed c-fos mRNA in rats treated with haloperidol (1 mg/kg, i.p.) Haloperidol 117-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 7914659-6 1994 for 1 h. Colocalization of c-fos mRNA in haloperidol-responsive NT neurons in the DLSt suggests that haloperidol"s induction of NT/N gene transcription may involve participation of the transcription factor Fos and the AP-1 consensus sequence in regulatory region of the NT/N gene. Haloperidol 41-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 7914659-6 1994 for 1 h. Colocalization of c-fos mRNA in haloperidol-responsive NT neurons in the DLSt suggests that haloperidol"s induction of NT/N gene transcription may involve participation of the transcription factor Fos and the AP-1 consensus sequence in regulatory region of the NT/N gene. Haloperidol 41-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 206-209 7914659-6 1994 for 1 h. Colocalization of c-fos mRNA in haloperidol-responsive NT neurons in the DLSt suggests that haloperidol"s induction of NT/N gene transcription may involve participation of the transcription factor Fos and the AP-1 consensus sequence in regulatory region of the NT/N gene. Haloperidol 101-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 7914659-6 1994 for 1 h. Colocalization of c-fos mRNA in haloperidol-responsive NT neurons in the DLSt suggests that haloperidol"s induction of NT/N gene transcription may involve participation of the transcription factor Fos and the AP-1 consensus sequence in regulatory region of the NT/N gene. Haloperidol 101-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 206-209 10465011-7 1994 A large peak in fos expression was seen at 0.5-2 h but only with the cytotoxic and regenerative proliferative treatments partial hepatectomy or carbon tetrachloride. Carbon Tetrachloride 144-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 8055340-1 1994 The GABA agonist muscimol, injected into the depressor area of the caudal ventrolateral medulla, increased blood pressure and increased the expression of the immediate early gene c-fos in the rostral ventral medulla (RVM) of the rat. Muscimol 17-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-184 8055340-2 1994 The number of Fos-immunoreactive (Fos-IR) neurons seen in the RVM was increased 3-fold after muscimol compared to Fos-IR after vehicle treatment. Muscimol 93-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 8055340-2 1994 The number of Fos-immunoreactive (Fos-IR) neurons seen in the RVM was increased 3-fold after muscimol compared to Fos-IR after vehicle treatment. Muscimol 93-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 8055340-2 1994 The number of Fos-immunoreactive (Fos-IR) neurons seen in the RVM was increased 3-fold after muscimol compared to Fos-IR after vehicle treatment. Muscimol 93-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 8055340-5 1994 Fos-IR neurons were also identified in the intermediolateral cell column of the thoracic spinal cord after muscimol injection, but were rarely observed in this area after vehicle treatment. Muscimol 107-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 8055341-5 1994 In addition, we assessed the ability of 2,5-AM (300 and 500 mg/kg) to induce Fos-like immunoreactivity (Fos-li) in the brain in sham-operated (SHAM), hepatic branch vagotomized (HBV) and total subdiaphragmatic vagotomized (TSDV) rats. 2,5-anhydromannitol 40-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-80 8055341-5 1994 In addition, we assessed the ability of 2,5-AM (300 and 500 mg/kg) to induce Fos-like immunoreactivity (Fos-li) in the brain in sham-operated (SHAM), hepatic branch vagotomized (HBV) and total subdiaphragmatic vagotomized (TSDV) rats. 2,5-anhydromannitol 40-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 8055341-6 1994 Both doses of 2,5-AM, but not control solutions, induced Fos-li in the area postrema (AP), nucleus of the solitary tract (NTS) and lateral parabrachial nucleus (1PBN). 2,5-anhydromannitol 14-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 8055341-8 1994 The effect of the lower 2,5-AM dose on Fos-li was blocked by HBV. 2,5-anhydromannitol 24-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 8084508-0 1994 Increased c-fos expression in nucleus of the solitary tract correlated with conditioned taste aversion to sucrose in rats. Sucrose 106-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-15 8075829-0 1994 Inhibition of c-Fos protein expression in rat spinal cord by antisense oligodeoxynucleotide superfusion. Oligodeoxyribonucleotides 71-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 8182549-0 1994 Induction of neurotensin and c-fos mRNA in distinct subregions of rat neostriatum after acute methamphetamine: comparison with acute haloperidol effects. Methamphetamine 94-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 8182549-4 1994 Additionally, this induction of NT/N gene transcription in the dorsolateral striatum by haloperidol appears to involve participation of the immediate early gene, c-fos. Haloperidol 88-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 8182549-7 1994 Unlike haloperidol, methamphetamine increased NT/N mRNA expression in the periventricular dorsomedial quadrant of the striatum and induced c-fos mRNA primarily in the medial aspects of the central core of the neostriatum. Methamphetamine 20-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 7914358-0 1994 Reserpine increases Fos activity in the rat basal ganglia via a quinpirole-sensitive mechanism. Reserpine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 8024692-7 1994 At low concentrations (5-200 microM), 1,10-phenanthroline increased the expression of insulin-stimulated g33, c-fos, and Egr-1 mRNA. 1,10-phenanthroline 38-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 7914358-0 1994 Reserpine increases Fos activity in the rat basal ganglia via a quinpirole-sensitive mechanism. Quinpirole 64-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 7914358-5 1994 Because dopamine is believed to excite striatal neurons via D1 receptors and inhibit them via D2 receptors, we hypothesized that acute dopamine depletion would increase Fos activity in basal ganglia circuits normally inhibited by dopaminergic input to D2 receptors. Dopamine 8-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-172 7914358-5 1994 Because dopamine is believed to excite striatal neurons via D1 receptors and inhibit them via D2 receptors, we hypothesized that acute dopamine depletion would increase Fos activity in basal ganglia circuits normally inhibited by dopaminergic input to D2 receptors. Dopamine 135-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-172 7914358-7 1994 The brains of rats perfused 3 h after reserpine displayed numerous Fos-like immunoreactive nuclei in the striatum, entopeduncular nucleus, nucleus accumbens shell, and ventral pallidum, and sparse Fos-like immunoreactive nuclei in the globus pallidus and nucleus accumbens core. Reserpine 38-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 7914358-9 1994 In the 3-h paradigm, pretreatment with the selective D1 antagonist SCH 23390 did not change the pattern of Fos-like immunoreactivity; pretreatment with the selective D2 agonist quinpirole completely blocked increased Fos synthesis. Quinpirole 177-187 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 217-220 7914358-10 1994 Acute dopamine depletion, therefore, increases Fos activity in the basal ganglia by disinhibiting D2 circuits. Dopamine 6-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 7969881-6 1994 The baseline expression of the gene c-fos in the offspring of male alcoholic rats did not differ from the norm; however, a powerful increase in the expression of the gene c-fos did appear in response to the administration of 2 g/kg of ethanol, in the absence of this effect in the control. Ethanol 235-242 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 7969881-6 1994 The baseline expression of the gene c-fos in the offspring of male alcoholic rats did not differ from the norm; however, a powerful increase in the expression of the gene c-fos did appear in response to the administration of 2 g/kg of ethanol, in the absence of this effect in the control. Ethanol 235-242 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-176 8032952-5 1994 Combined demonstration in the spinal cord of Fos protein-like immunoreactive neurons and neurons labeled retrogradely with Fluoro-Gold from the gracile nucleus showed that some of the Fos protein-like immunoreactive neurons in Rexed"s laminae III and IV contributed to the postsynaptic dorsal column pathway. 2-hydroxy-4,4'-diamidinostilbene, methanesulfonate salt 123-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 184-187 7519273-6 1994 Within 30 min after Ipt administration, the c-fos gene was upregulated by 10-fold, and thereafter, its message level decreased and stabilized at approximately 3-fold over control. Isoproterenol 20-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 8032900-0 1994 Changes in c-fos induction in dorsal horn neurons by hindpaw formalin stimulation following tibial neurotomy. Formaldehyde 61-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 7915411-6 1994 In normally developing rats, amphetamine induced Fos expression in both the striatum and globus pallidus by two weeks after birth; by four weeks, the pattern of amphetamine-induced Fos immunoreactivity was similar to that observed in adults. Amphetamine 29-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 7915411-6 1994 In normally developing rats, amphetamine induced Fos expression in both the striatum and globus pallidus by two weeks after birth; by four weeks, the pattern of amphetamine-induced Fos immunoreactivity was similar to that observed in adults. Amphetamine 29-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 181-184 7915411-6 1994 In normally developing rats, amphetamine induced Fos expression in both the striatum and globus pallidus by two weeks after birth; by four weeks, the pattern of amphetamine-induced Fos immunoreactivity was similar to that observed in adults. Amphetamine 161-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 7915411-6 1994 In normally developing rats, amphetamine induced Fos expression in both the striatum and globus pallidus by two weeks after birth; by four weeks, the pattern of amphetamine-induced Fos immunoreactivity was similar to that observed in adults. Amphetamine 161-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 181-184 7915411-7 1994 In the globus pallidus of both two- and three-week-old rats, amphetamine induced greater expression of Fos than in adults. Amphetamine 61-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-106 7915411-9 1994 In striatal grafts, amphetamine induced Fos expression from three weeks after implantation onwards, and by five to 10 weeks post-grafting the pattern of Fos immunoreactivity was similar to that observed in adult grafts. Amphetamine 20-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 7915411-9 1994 In striatal grafts, amphetamine induced Fos expression from three weeks after implantation onwards, and by five to 10 weeks post-grafting the pattern of Fos immunoreactivity was similar to that observed in adult grafts. Amphetamine 20-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-156 7915411-10 1994 However, apomorphine induced a considerable number of Fos-positive nuclei in striatal grafts at three and four weeks after grafting. Apomorphine 9-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 8184987-2 1994 In six male Wistar-Kyoto rats (WKY), unilateral injection of an antisense oligonucleotide to c-fos mRNA suppressed the expression of Fos-like immunoreactivity in neurons in the RVM in response to inhibition of depressor neurons in the caudal ventrolateral medulla (CVLM). Oligonucleotides 74-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 8184987-2 1994 In six male Wistar-Kyoto rats (WKY), unilateral injection of an antisense oligonucleotide to c-fos mRNA suppressed the expression of Fos-like immunoreactivity in neurons in the RVM in response to inhibition of depressor neurons in the caudal ventrolateral medulla (CVLM). Oligonucleotides 74-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-136 8184987-5 1994 These studies demonstrate that expression of c-fos in the RVM can be blocked in vivo by treatment with an antisense oligonucleotide, and that basal and stimulated expression of the c-fos gene is important in the central control of arterial blood pressure. Oligonucleotides 116-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 7913201-0 1994 Nicotine induced c-fos expression in the striatum is mediated mostly by dopamine D1 receptor and is dependent on NMDA stimulation. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 7913201-0 1994 Nicotine induced c-fos expression in the striatum is mediated mostly by dopamine D1 receptor and is dependent on NMDA stimulation. N-Methylaspartate 113-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 7913201-2 1994 To extend our understanding of nicotine and dopamine interactions, we mapped the pattern of c-fos expression in the striatum as an important marker of some of the earliest changes that occur at gene transcription level. Nicotine 31-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 7913201-3 1994 Acute nicotine injections in rats led to Fos expression more prominently in the caudatoputamen than in the nucleus accumbens in a dose-dependent fashion. Nicotine 6-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 7913201-5 1994 Injections of mecamylamine completely blocked nicotine-induced Fos expression. Mecamylamine 14-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 7913201-5 1994 Injections of mecamylamine completely blocked nicotine-induced Fos expression. Nicotine 46-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 7913201-6 1994 Injections of the selective dopamine D1 antagonist SCH 23390, but not D2 antagonist YM 09151-2 or Clozapine, a drug with high affinity to D4 receptors, before nicotine injections, completely blocked Fos expression in the striatum. Dopamine 28-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-202 7913201-7 1994 Nicotine induced Fos expression was also blocked completely by the NMDA receptor antagonists MK-801 and CPP. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 7913201-7 1994 Nicotine induced Fos expression was also blocked completely by the NMDA receptor antagonists MK-801 and CPP. Dizocilpine Maleate 93-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 7913201-8 1994 These results suggest that nicotine-induced Fos expression in the striatum is mediated mostly by dopamine D1 receptors and that the Fos expression is also dependent on N-methyl-D-aspartate (NMDA) stimulation. Nicotine 27-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 7913201-8 1994 These results suggest that nicotine-induced Fos expression in the striatum is mediated mostly by dopamine D1 receptors and that the Fos expression is also dependent on N-methyl-D-aspartate (NMDA) stimulation. N-Methylaspartate 168-188 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-135 7913201-8 1994 These results suggest that nicotine-induced Fos expression in the striatum is mediated mostly by dopamine D1 receptors and that the Fos expression is also dependent on N-methyl-D-aspartate (NMDA) stimulation. N-Methylaspartate 190-194 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-135 8028480-1 1994 Acute administration of the typical neuroleptic haloperidol (HAL, 2 mg/kg) induced the immediate-early gene proteins (IEGPs) c-Fos, Fos-related antigens (FRAs), FosB, JunB, JunD and Krox24 in the striatum and nucleus accumbens of the rat brain. Haloperidol 48-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 8028480-1 1994 Acute administration of the typical neuroleptic haloperidol (HAL, 2 mg/kg) induced the immediate-early gene proteins (IEGPs) c-Fos, Fos-related antigens (FRAs), FosB, JunB, JunD and Krox24 in the striatum and nucleus accumbens of the rat brain. Haloperidol 48-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-130 8028480-1 1994 Acute administration of the typical neuroleptic haloperidol (HAL, 2 mg/kg) induced the immediate-early gene proteins (IEGPs) c-Fos, Fos-related antigens (FRAs), FosB, JunB, JunD and Krox24 in the striatum and nucleus accumbens of the rat brain. Haloperidol 61-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 8028480-1 1994 Acute administration of the typical neuroleptic haloperidol (HAL, 2 mg/kg) induced the immediate-early gene proteins (IEGPs) c-Fos, Fos-related antigens (FRAs), FosB, JunB, JunD and Krox24 in the striatum and nucleus accumbens of the rat brain. Haloperidol 61-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-130 8028480-2 1994 In contrast, acute administration of the atypical antipsychotic drug clozapine (CLOZ, 30 mg/kg) induced only FRAs, JunB and Krox24 IEGPs in the striatum, and c-Fos, FRAs, and Krox24 IEGPs in the nucleus accumbens. Clozapine 69-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 8028480-6 1994 Specifically, CLOZ induces FRAs in the islands of Calleja and lateral septum and this action may be involved in its therapeutic effects on the negative symptoms of schizophrenia, whereas HAL produces a coordinate induction of Fos and JunB in striatal neurons and this dimer combination may be involved in producing the extrapyramidal side-effects of typical neuroleptics. Haloperidol 187-190 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 226-229 7942088-2 1994 Although nicotine has been reported to induce c-fos, in the present study it was shown that this induction does not alter the accumulation of a number of transcripts associated with AD. Nicotine 9-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 8049720-4 1994 We found that over 80% of the VCS-induced Fos-IR neurons in the medial division of the bed nucleus of the stria terminalis also contained estrogen receptor-immunoreactivity. VCP 30-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 8019941-6 1994 Our data suggest that fos, myc, ras and jun protooncogenes were expressed from 15" to 48 h after treatment with either estrogen or tamoxifen. Tamoxifen 131-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-25 8032900-3 1994 The excitability of dorsal horn neurons was expressed as the percentage ratio of the number of formalin-induced c-fos protein-like immunoreactive neurons (fos-neurons) on the neurotomized (experimental) side to that on the un-neurotomized (control) side. Formaldehyde 95-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 8032680-10 1994 TRH-induced jun B and c-fos responses were characteristic of immediate early genes as shown by the superinduction observed under cycloheximide treatment and the total inhibition elicited by actinomycin D. Cycloheximide 129-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 8032680-10 1994 TRH-induced jun B and c-fos responses were characteristic of immediate early genes as shown by the superinduction observed under cycloheximide treatment and the total inhibition elicited by actinomycin D. Dactinomycin 190-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 8065795-0 1994 Chronic treatments with aspirin or acetaminophen reduce both the development of polyarthritis and Fos-like immunoreactivity in rat lumbar spinal cord. Aspirin 24-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 8065795-0 1994 Chronic treatments with aspirin or acetaminophen reduce both the development of polyarthritis and Fos-like immunoreactivity in rat lumbar spinal cord. Acetaminophen 35-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 8065795-2 1994 The aim of this study was to evaluate the suitability of the Fos-LI technique to gauge the effects of the two most prescribed analgesics, aspirin and acetaminophen (paracetamol), on spinal cord neurons of polyarthritic rats. Aspirin 138-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 8065795-2 1994 The aim of this study was to evaluate the suitability of the Fos-LI technique to gauge the effects of the two most prescribed analgesics, aspirin and acetaminophen (paracetamol), on spinal cord neurons of polyarthritic rats. Acetaminophen 150-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 8065795-2 1994 The aim of this study was to evaluate the suitability of the Fos-LI technique to gauge the effects of the two most prescribed analgesics, aspirin and acetaminophen (paracetamol), on spinal cord neurons of polyarthritic rats. Acetaminophen 165-176 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 8065795-7 1994 (2) Despite the fact that the clinical signs of arthritis were reduced, basal Fos-LI induced by AIA disease was not changed after a 2-week chronic treatment with either aspirin (300 mg/kg/day) or acetaminophen (500 mg/kg/day) starting 3 weeks after AIA inoculation, i.e., at the maximal stage of hyperalgesia and when Fos-LI is maximal. aia 96-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 8065795-9 1994 (3) In contrast, when the same chronic treatment was applied during the development of AIA, i.e., 1 week after inoculation, the number of Fos-LI nuclei was significantly decreased (about 50%) in aspirin- and acetaminophen-treated groups as compared to vehicle-treated groups. Aspirin 195-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-141 8065795-9 1994 (3) In contrast, when the same chronic treatment was applied during the development of AIA, i.e., 1 week after inoculation, the number of Fos-LI nuclei was significantly decreased (about 50%) in aspirin- and acetaminophen-treated groups as compared to vehicle-treated groups. Acetaminophen 208-221 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-141 7827709-3 1994 Even minute stimuli such as the attachment of electrodes, needle injection and saline administration increase the level of c-fos mRNA expression in animal brains. Sodium Chloride 79-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 7827709-7 1994 Rough handling with repeated saline administration enhanced cortical c-fos mRNA expression in the rat brain after a single saline injection by increasing baseline c-fos mRNA levels. Sodium Chloride 29-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 7827709-7 1994 Rough handling with repeated saline administration enhanced cortical c-fos mRNA expression in the rat brain after a single saline injection by increasing baseline c-fos mRNA levels. Sodium Chloride 29-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-168 7827709-7 1994 Rough handling with repeated saline administration enhanced cortical c-fos mRNA expression in the rat brain after a single saline injection by increasing baseline c-fos mRNA levels. Sodium Chloride 123-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 7827709-8 1994 In contrast, gentle handling with repeated saline administration diminished c-fos mRNA expression after a single injection by decreasing baseline c-fos mRNA levels. Sodium Chloride 43-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 7827709-8 1994 In contrast, gentle handling with repeated saline administration diminished c-fos mRNA expression after a single injection by decreasing baseline c-fos mRNA levels. Sodium Chloride 43-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-151 8019836-0 1994 2-Mercaptoacetate and 2-deoxy-D-glucose induce Fos-like immunoreactivity in rat brain. 2-mercaptoacetate 0-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 8019836-0 1994 2-Mercaptoacetate and 2-deoxy-D-glucose induce Fos-like immunoreactivity in rat brain. Deoxyglucose 22-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 8019836-3 1994 Remote intravenous infusions of both MA and 2-DG induced Fos-like immunoreactivity (Fos-li) in specific brain sites, but the pattern was different for the two drugs. Deoxyglucose 44-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 8019836-3 1994 Remote intravenous infusions of both MA and 2-DG induced Fos-like immunoreactivity (Fos-li) in specific brain sites, but the pattern was different for the two drugs. Deoxyglucose 44-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 8019836-4 1994 Mercaptoacetate induced Fos-li in the nucleus of the solitary tract (NTS), the central subnucleus of the lateral parabrachial nucleus (1PBN), the central nucleus of the amygdala (CNA, lateral part) and the dorsal motor nucleus of the vagus (DMV). Thioglycolates 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 8019836-6 1994 2-Deoxy-D-glucose also induced Fos-li in the NTS, CNA (lateral part) and DMV, as well as in the external 1PBN subnucleus, locus coeruleus, paraventricular and supraoptic hypothalamic nuclei, and in scattered cells throughout the diencephalon. Deoxyglucose 0-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 8019836-6 1994 2-Deoxy-D-glucose also induced Fos-li in the NTS, CNA (lateral part) and DMV, as well as in the external 1PBN subnucleus, locus coeruleus, paraventricular and supraoptic hypothalamic nuclei, and in scattered cells throughout the diencephalon. (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 73-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 8019836-7 1994 Induction of Fos-li by 2-DG was not blocked by vagotomy. Deoxyglucose 23-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 8019836-8 1994 Results suggest that 2-DG"s effects on Fos-li are mediated by a direct central action, whereas MA"s effects are mediated by peripheral sensory neurons. Deoxyglucose 21-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 8019836-10 1994 The correlation of Fos-immunoreactive sites with sites where lesions have been shown to cause deficits in MA- and 2-DG-induced feeding indicates that c-fos expression defines in part the central pathways involved in the metabolic control of feeding. Deoxyglucose 114-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 8019836-10 1994 The correlation of Fos-immunoreactive sites with sites where lesions have been shown to cause deficits in MA- and 2-DG-induced feeding indicates that c-fos expression defines in part the central pathways involved in the metabolic control of feeding. Deoxyglucose 114-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-155 8019847-0 1994 Water-avoidance stress-induced c-fos expression in the rat brain and stimulation of fecal output: role of corticotropin-releasing factor. Water 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 8019847-1 1994 Immunohistochemical detection of the immediate-early gene c-fos was used to determine the pattern of neuronal activity in the rat brain after exposure to water-avoidance stress known to stimulate fecal output in rats. Water 154-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 8019847-2 1994 Avoidance to water for 1 h by standing on a small platform increases pellet output and induces numerous Fos-positive cells in the parvocellular part of the paraventricular nucleus of the hypothalamus (PVN), locus coeruleus (LC) and, to a lesser extent, in the bed nucleus of the stria terminalis, lateral septum, dorsal raphe nucleus and A5 and A1 noradrenergic neurons. Water 13-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 7520134-0 1994 Differences in the regional and cellular localization of c-fos messenger RNA induced by amphetamine, cocaine and caffeine in the rat. Amphetamine 88-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 7520134-0 1994 Differences in the regional and cellular localization of c-fos messenger RNA induced by amphetamine, cocaine and caffeine in the rat. Cocaine 101-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 7520134-0 1994 Differences in the regional and cellular localization of c-fos messenger RNA induced by amphetamine, cocaine and caffeine in the rat. Caffeine 113-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 7520134-5 1994 All drug treatments except 3 mg/kg cocaine induced an increased level of c-fos messenger RNA in cells that had a neuron-like morphology. Cocaine 35-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 7520134-12 1994 With cocaine treatment (30 mg/kg), about 30% of striatal neuron-like cells were c-fos labelled. Cocaine 5-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 7520134-17 1994 With caffeine treatment, about 73% of neuron-like cells were c-fos labelled in the lateral striatum, but labelling was much less pronounced in the medial part or in the accumbens. Caffeine 5-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 8004454-0 1994 Reversal of dexfenfluramine-induced anorexia and c-Fos/c-Jun expression by lesion in the lateral parabrachial nucleus. Dexfenfluramine 12-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 7510676-11 1994 One consequence of the activation of STAT91 in the nucleus is the appearance of GH-stimulated DNA binding activity, as assessed by gel-mobility shift assay using an oligonucleotide containing a c-sis-inducible element from the c-fos promoter. Oligonucleotides 165-180 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 227-232 8019847-4 1994 reduced water-avoidance stress-induced c-fos expression mainly in the PVN and the LC (44 and 60%, respectively) and decreased by 60% the stimulated fecal output. Water 8-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 7516994-0 1994 Expression of nitric oxide synthase and colocalisation with Jun, Fos and Krox transcription factors in spinal cord neurons following noxious stimulation of the rat hindpaw. Nitric Oxide 14-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 7516994-11 1994 Double-staining also demonstrated that many Jun, Fos and Krox labelled neurons are in close proximity to NDP labelled nerve fibers suggesting a functional relationship between expression of immediate-early gene encoded transcription factors and presence of nitric oxide in the rat spinal cord. Nitric Oxide 257-269 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 8008200-7 1994 The number of Fos-like immunoreactive neurons induced by the cold stimulation temperatures was significantly decreased by pretreatment with 10 mg/kg s.c. morphine and moderately decreased by 5 mg/kg s.c. Morphine 154-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 8008200-9 1994 Naloxone (2 mg/kg s.c.) significantly increased the number of Fos-like immunoreactive neurons induced by -20 degrees C as compared to saline-injected rats. Naloxone 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 8025533-7 1994 When given 30 min prior to the injury, MK-801 (4 mg/kg) prevented the increased expression of c-fos but did not affect the later rise in procholecystokinin-mRNA. Dizocilpine Maleate 39-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 8022627-5 1994 Placing the tail in 50 degrees C water before each hindpaw pinch significantly reduced Fos-like immunoreactivity in these regions. Water 33-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 7910104-1 1994 Pretreatment of rats for 4 days with the selective dopamine D1 receptor agonist A-77636 attenuated the ability of cocaine to induce locomotor hyperactivity and to stimulate the expression of the proto-oncogene c-fos in the striatum and nucleus accumbens. Cocaine 114-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-215 8974322-1 1994 The expression of the proteins (C-FOS and C-JUN) encoded by the immediate early genes c-fos and c-jun was investigated in the brains of rats undergoing ethanol withdrawal. Ethanol 152-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 8016407-0 1994 Cholecystokinin- and dexfenfluramine-induced anorexia compared using devazepide and c-fos expression in the rat brain. Dexfenfluramine 21-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 8016407-2 1994 The present study compared the patterns of c-fos protein-like immunoreactivity (FLI) induced in rat brain by CCK and the indirect 5HT agonist dexfenfluramine (DFEN), as well as the ability for devazepide, a CCK-A receptor antagonist, to antagonize both anorexia and FLI induced by these agents. Dexfenfluramine 142-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 8012803-6 1994 LiCl administration, a common unconditioned stimulus (UCS) in CTA experiments, was found to induce c-Fos protein in a number of brainstem regions, including the nucleus of the solitary tract (NTS), medial and lateral pontine parabrachial nucleus (PBN), and hypoglossal nucleus. Lithium Chloride 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 8177522-1 1994 We studied the ability of the rat vasoactive intestinal polypeptide (VIP) cyclic AMP responsive element (CRE) to regulate reporter gene expression through a c-fos promoter in rat sensory neurons transfected in culture by plasmid microinjection. Cyclic AMP 74-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-162 8141390-2 1994 After hydralazine-induced hypotension or sinoaortic denervation, two treatments that reduce baroreceptor afferent activity, numerous Fos-positive neurons were observed in the RVLM. Hydralazine 6-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-136 8141390-3 1994 The number of Fos-positive neurons in the RVLM was counted in brain stem sections from hydralazine-treated rats that had been previously injected with Fluorogold into the upper thoracic spinal cord to label spinally projecting RVLM neurons as well as stained for phenylethanolamine-N-methyltransferase (PNMT) as a marker of C1 neurons. Hydralazine 87-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 8141390-4 1994 The results indicate that approximately 80% of the C1 neurons expressed Fos in response to hydralazine injection; this was true of spinally projecting C1 neurons as well as those C1 neurons that were not labeled with Fluorogold. Hydralazine 91-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 8141390-5 1994 Furthermore, in hydralazine-treated rats, the majority of Fluorogold-labeled Fos-positive neurons contained PNMT. Hydralazine 16-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-80 8170349-8 1994 Furthermore, prevention of c-fos translation by pretreatment with protein synthesis inhibitor cycloheximide attenuated this dehydration induced increase in vasopressin mRNA. Cycloheximide 94-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 8294911-0 1994 Stimulation of the cyclic GMP pathway by NO induces expression of the immediate early genes c-fos and junB in PC12 cells. Cyclic GMP 19-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 8294911-4 1994 We found that expression of c-fos and junB but not of c-jun or junD is increased upon activation of cyclic GMP pathway. Cyclic GMP 100-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 8190362-0 1994 Induction of c-fos mRNA expression in an in vitro hippocampal slice model of adult rats after kainate but not gamma-aminobutyric acid or bicuculline treatment. Kainic Acid 94-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 8190362-2 1994 Following a short-term exposure to kainate, c-fos gene expression was induced by 12-fold in the adult, but not the newborn, hippocampus. Kainic Acid 35-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 8137158-0 1994 Dopamine agonists and stress produce different patterns of Fos-like immunoreactivity in the lateral habenula. Dopamine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 8137158-1 1994 In rats treated systemically with either amphetamine, amfonelic acid or apomorphine, large numbers of cells displaying Fos-like immunoreactivity (FLI) could be seen in the lateral zone of the lateral habenula. Amphetamine 41-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-122 8137158-1 1994 In rats treated systemically with either amphetamine, amfonelic acid or apomorphine, large numbers of cells displaying Fos-like immunoreactivity (FLI) could be seen in the lateral zone of the lateral habenula. amfonelic acid 54-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-122 8137158-1 1994 In rats treated systemically with either amphetamine, amfonelic acid or apomorphine, large numbers of cells displaying Fos-like immunoreactivity (FLI) could be seen in the lateral zone of the lateral habenula. Apomorphine 72-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-122 8276812-7 1994 This serine/threonine kinase, termed Fos kinase, has been purified > 24,000-fold through five column steps to near homogeneity and is shown to be a 37-kDa protein as determined by SDS-polyacrylamide gel electrophoresis (PAGE) with a pI = 6.0. Sodium Dodecyl Sulfate 183-186 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 8276812-7 1994 This serine/threonine kinase, termed Fos kinase, has been purified > 24,000-fold through five column steps to near homogeneity and is shown to be a 37-kDa protein as determined by SDS-polyacrylamide gel electrophoresis (PAGE) with a pI = 6.0. polyacrylamide 187-201 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 8276812-12 1994 Moreover, Fos kinase phosphorylation of c-Fos in vitro results in a similar electrophoretic mobility shift, demonstrating that Fos kinase may be responsible for growth factor-stimulated alterations in mobility on SDS-PAGE and phosphorylation of this transcription factor. Sodium Dodecyl Sulfate 213-216 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-13 7889209-5 1994 CV-11974, an antagonist of ANG II receptor type-1 (AT1), inhibited the ANG II-induced expression of c-fos, c-myc and ET-1 mRNAs, as well as that of ETB receptor mRNA, whereas PD-123319, an antagonist of the ANG II type-2 (AT2) receptor, failed to block such induction. candesartan 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 8193923-7 1994 The c-fos-LI was effectively reduced by preadministration of the N-methyl-D-aspartate (NMDA) receptor antagonist, ketamine (100 mg/kg, IP). Ketamine 114-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 8193936-3 1994 The cyclooxygenase inhibitor indomethacin and the glutamate NMDA antagonist MK801 inhibited c-fos protein in the paraventricular nucleus (PVN), supraoptic nucleus (SON), and the A1/A2 regions of the brain stem induced by IP or IV injections of LPS (40 micrograms). Indomethacin 29-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 8193936-3 1994 The cyclooxygenase inhibitor indomethacin and the glutamate NMDA antagonist MK801 inhibited c-fos protein in the paraventricular nucleus (PVN), supraoptic nucleus (SON), and the A1/A2 regions of the brain stem induced by IP or IV injections of LPS (40 micrograms). Glutamic Acid 50-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 8193936-3 1994 The cyclooxygenase inhibitor indomethacin and the glutamate NMDA antagonist MK801 inhibited c-fos protein in the paraventricular nucleus (PVN), supraoptic nucleus (SON), and the A1/A2 regions of the brain stem induced by IP or IV injections of LPS (40 micrograms). N-Methylaspartate 60-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 8193936-3 1994 The cyclooxygenase inhibitor indomethacin and the glutamate NMDA antagonist MK801 inhibited c-fos protein in the paraventricular nucleus (PVN), supraoptic nucleus (SON), and the A1/A2 regions of the brain stem induced by IP or IV injections of LPS (40 micrograms). Dizocilpine Maleate 76-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 8193936-4 1994 The H1 histamine antagonist diphenhydramine, but not the H2 histamine antagonist cimetidine, reduced the amount of c-fos labeling. Histamine 7-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 8193936-4 1994 The H1 histamine antagonist diphenhydramine, but not the H2 histamine antagonist cimetidine, reduced the amount of c-fos labeling. Diphenhydramine 28-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 8193936-5 1994 MK801 also attenuated the effects of stress (foot shock) on c-fos protein; however, indomethacin had no effect on c-fos protein induced by stress. Dizocilpine Maleate 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 8055349-0 1994 Protective effect of bifemelane on c-Fos-like immunoreactivity in rat cerebral ischemia. bifemelane 21-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 8055349-4 1994 In animals that had been injected with bifemelane hydrochloride (20 mg/kg, IP) 30 min before the onset of ischemia and 90 min after reperfusion, the number of c-Fos-like immunoreactive neurons was significantly reduced in the cerebral cortex. bifemelane 39-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-164 8055349-5 1994 The results suggest that bifemelane hydrochloride can inhibit the ischemia-induced increase in c-Fos-like immunoreactivity in cerebral cortex neurons. bifemelane 25-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 8087422-3 1994 Previous work in our laboratory demonstrated by gel shift analysis that Fos and non-Fos-containing complexes formed with oligonucleotides containing this element. Oligonucleotides 121-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 8087422-3 1994 Previous work in our laboratory demonstrated by gel shift analysis that Fos and non-Fos-containing complexes formed with oligonucleotides containing this element. Oligonucleotides 121-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 8286384-8 1994 264, 3538-3544], chronic (24 h) treatment with bryostatin blocked the formation of neurites in response to NGF or basic fibroblast-derived growth factor stimulation, but, like PMA, bryostatin did not block the induction of c-fos or c-jun protooncogenes by NGF. Bryostatins 47-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 223-228 8276812-12 1994 Moreover, Fos kinase phosphorylation of c-Fos in vitro results in a similar electrophoretic mobility shift, demonstrating that Fos kinase may be responsible for growth factor-stimulated alterations in mobility on SDS-PAGE and phosphorylation of this transcription factor. Sodium Dodecyl Sulfate 213-216 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 8276812-12 1994 Moreover, Fos kinase phosphorylation of c-Fos in vitro results in a similar electrophoretic mobility shift, demonstrating that Fos kinase may be responsible for growth factor-stimulated alterations in mobility on SDS-PAGE and phosphorylation of this transcription factor. Sodium Dodecyl Sulfate 213-216 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 8304461-5 1994 Studies using the protein synthesis inhibitor cycloheximide suggest that c-fos and c-jun are part of the "immediate-early" response of the small intestine. Cycloheximide 46-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 8193936-8 1994 Thus, prostaglandin synthesis, glutamate release, histamine receptors, and visceral afferents represent functional biochemical and neural pathways through which endotoxin activates c-fos protein in specific autonomic and neuroendocrine regulatory nuclei. Prostaglandins 6-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 181-186 7914492-2 1994 Kainic acid injections resulted in a widespread pattern of Fos protein induction, mainly involving cortical olfactory structures and hippocampus. Kainic Acid 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 7914492-3 1994 Immunoreactive cells were observed in large number 2-24 h after injection and had almost completely disappeared by 48 h. NMDA injections elicited a shorter (2-8 h) expression of Fos protein, involving a lower number of cells in cortical olfactory structures, a much larger number of cells in the other cortical regions, and not involving the hippocampus at all. N-Methylaspartate 121-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 178-181 7914492-7 1994 The expression of Fos protein, however, was partially prevented only in NMDA cases. N-Methylaspartate 72-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 8297482-4 1994 Quantitative northern blot analysis of mRNA was used to examine the expression of the oncogenes myc, fos, and Ha-ras in the livers of animals treated with furan. furan 155-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-104 7946406-5 1994 Inhibition of cell growth by butyrate was associated with a complex pattern of cell cycle regulated gene expression, including a decoupling of c-fos and c-jun gene expression. Butyrates 29-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-148 7946406-6 1994 Transcription of c-fos, as well as c-jun increased with butyrate arrest, whereas steady rate mRNA levels of c-jun only were increased, suggesting additional regulation of c-fos. Butyrates 56-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 8263051-0 1994 Prostaglandin E1 induces c-Fos and Myc proteins and protects rat hippocampal cells against hypoxic injury. Alprostadil 0-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 8263051-7 1994 Western blot analysis showed an increased induction of 62-kDa c-Fos and 58, 60, and 66 kDa Myc proteins in rat hippocampal cells with 10(-7) M PGE1 treatment. Alprostadil 143-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 8297482-5 1994 In male rats, a single administration of 30 mg/kg furan produced necrosis and a subsequent wave of cell proliferation 48 h after treatment and induced transient peaks in the expression of myc, fos, and Ha-ras 6-24 h after treatment. furan 50-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-196 7700853-0 1994 Expression of the FOS proto-oncogene protein in brain after ICV administration of Tyr-W-MIF-1 (Tyr-Pro-Trp-Gly-NH2). Tyrosine 82-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 8152541-0 1994 Effects of descending inhibitory systems on the c-Fos expression in the rat spinal cord during formalin-induced noxious stimulation. Formaldehyde 95-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 8152542-0 1994 Application of morphine prior to noxious stimulation differentially modulates expression of Fos, Jun and Krox-24 proteins in rat spinal cord neurons. Morphine 15-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 8152542-8 1994 At 2 h following noxious heat stimulation morphine had decreased the number of labelled neurons for c-Fos, Fos B, Krox-24, c-Jun and Jun B to 30-60% of control levels in laminae I-II and to 10-30% in laminae III-VII,X of the spinal cord. Morphine 42-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 7700853-0 1994 Expression of the FOS proto-oncogene protein in brain after ICV administration of Tyr-W-MIF-1 (Tyr-Pro-Trp-Gly-NH2). Tyrosine 95-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 7700853-0 1994 Expression of the FOS proto-oncogene protein in brain after ICV administration of Tyr-W-MIF-1 (Tyr-Pro-Trp-Gly-NH2). Glycine 107-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 7700853-8 1994 The results show that Tyr-W-MIF-1 induces FOS activation in several brain areas, including but not limited to, areas associated with nociception and stress-induced analgesia. Tyrosine 22-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 8298998-2 1993 Augmentation of c-fos and NGFI-A mRNA by light is apparently associated with activation of cholinergic nicotinic and muscarinic receptors as it can be suppressed by the nicotinic antagonist mecamylamine and the muscarinic antagonist atropine. Mecamylamine 190-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 8159434-2 1994 Intravenous injection of Kelatorphan (5, 10 and 20 mg/kg), an inhibitor of multiple enkephalin-degrading enzymes, 20 min before noxious stimulation reduced the overall number of dorsal horn neurons expressing c-FOS and NGF1-A by up to 20-30%. kelatorphan 25-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 209-214 8159434-4 1994 Morphine (5, 7.5 and 10 mg/kg) produced a dose-dependent reduction of c-FOS expression by up to 70% only when injected before noxious stimulation. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 8159434-7 1994 Naloxone itself slightly increased the overall number of c-FOS-positive neurons in all laminae of the spinal cord. Naloxone 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 21551750-0 1994 Fos-like immunoreactivity in the rat spinal cord induced by formalin injection in the forelimb to gauge possible plasticity of primary afferent fibers following partial deafferentation. Formaldehyde 60-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 21551750-5 1994 In control animals, following the formalin injection the greatest number of Fos-LI neurons was encountered in the superficial laminae and in the neck of the dorsal horn of segments C5-T1. Formaldehyde 34-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 8307126-0 1994 Neural substrate of a cerebellar movement disorder induced by intracerebroventricular injection of propidium iodide in the rat: a Fos immunocytochemical study. Propidium 99-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-133 7778409-6 1994 In adult rats that had been treated neonatally with capsaicin, there was a marked fall in c-fos activation by mechanical or chemical noxious stimuli in all immunoreactive areas. Capsaicin 52-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 8010084-0 1994 Topical capsaicin treatment suppresses formalin-induced fos expression in rat spinal cord. Capsaicin 8-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 8010084-0 1994 Topical capsaicin treatment suppresses formalin-induced fos expression in rat spinal cord. Formaldehyde 39-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 8010084-2 1994 The present study was undertaken to investigate the effects of topical application of Cap to sciatic nerve on the formalin-induced expression of proto-oncogene proteins c-fos in the rat spinal cord using immunohistochemical display of fos-like protein. Formaldehyde 114-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-174 8010084-2 1994 The present study was undertaken to investigate the effects of topical application of Cap to sciatic nerve on the formalin-induced expression of proto-oncogene proteins c-fos in the rat spinal cord using immunohistochemical display of fos-like protein. Formaldehyde 114-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-174 8010084-3 1994 In rats subjected to formalin injection into the hind paw, numerous fos-like immunoreactivity (FLI) neurons were found in the spinal dorsal horn, with heavy labeling in laminae I-II and V-VI. Formaldehyde 21-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 8010084-5 1994 It was suggested that activation of Cap-sensitive unmyelinated nociceptive afferents following formalin injection was primarily responsible for the activation of c-fos gene. Formaldehyde 95-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 7908600-1 1993 Fos expression in the globus pallidus (GP) of rats was elicited by the D2 agonist quinpirole both ipsilateral and contralateral to a unilateral nigrostriatal 6-hydroxydopamine (6-OHDA) injection; however, the 6-OHDA-treated hemisphere was more sensitive to this effect. Quinpirole 82-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 7908600-1 1993 Fos expression in the globus pallidus (GP) of rats was elicited by the D2 agonist quinpirole both ipsilateral and contralateral to a unilateral nigrostriatal 6-hydroxydopamine (6-OHDA) injection; however, the 6-OHDA-treated hemisphere was more sensitive to this effect. Oxidopamine 158-175 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 7908600-1 1993 Fos expression in the globus pallidus (GP) of rats was elicited by the D2 agonist quinpirole both ipsilateral and contralateral to a unilateral nigrostriatal 6-hydroxydopamine (6-OHDA) injection; however, the 6-OHDA-treated hemisphere was more sensitive to this effect. Oxidopamine 177-183 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 7908600-1 1993 Fos expression in the globus pallidus (GP) of rats was elicited by the D2 agonist quinpirole both ipsilateral and contralateral to a unilateral nigrostriatal 6-hydroxydopamine (6-OHDA) injection; however, the 6-OHDA-treated hemisphere was more sensitive to this effect. Oxidopamine 209-215 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 7908600-2 1993 The quinpirole-induced GP Fos expression was antagonized in both hemispheres by the D2 antagonist eticlopride, but not by the D1 antagonist SCH 23390. Quinpirole 4-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 7908600-2 1993 The quinpirole-induced GP Fos expression was antagonized in both hemispheres by the D2 antagonist eticlopride, but not by the D1 antagonist SCH 23390. eticlopride 98-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 7908600-3 1993 In neurologically intact rats, the D1 agonist SKF 38393, which alone did not elicit pallidal Fos expression, augmented the quinpirole-induced Fos response. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 46-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-145 7908600-3 1993 In neurologically intact rats, the D1 agonist SKF 38393, which alone did not elicit pallidal Fos expression, augmented the quinpirole-induced Fos response. Quinpirole 123-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-145 8149221-1 1993 To investigate the distribution of Fos-like immunoreactivity (FLI) in the central nervous system of urethane anesthesized rats after activation of a somatosympathetic reflex pathway, the cut central end of the right femoral nerve of 17 male Wistar rats was stimulated electrically for 1 h at parameters such that increases in heart rate and arterial pressure were elicited. Urethane 100-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 8010091-1 1994 C-fos proteins were visualized immunohistochemically in the brain of rats after seizures induced by injecting penicillin into hippocampus and by penicillin+electroacupuncture treatment. Penicillins 110-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8010091-1 1994 C-fos proteins were visualized immunohistochemically in the brain of rats after seizures induced by injecting penicillin into hippocampus and by penicillin+electroacupuncture treatment. Penicillins 145-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8298998-2 1993 Augmentation of c-fos and NGFI-A mRNA by light is apparently associated with activation of cholinergic nicotinic and muscarinic receptors as it can be suppressed by the nicotinic antagonist mecamylamine and the muscarinic antagonist atropine. Atropine 233-241 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 8298998-3 1993 Moreover, the light-induced increase of c-fos mRNA in retina appears to be associated with activation of glutamate receptors also as the noncompetitive inhibitor of N-methyl-D-aspartate receptors dizocilpine (MK-801) partially suppressed the increase of the c-fos message. Dizocilpine Maleate 196-207 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 8298998-3 1993 Moreover, the light-induced increase of c-fos mRNA in retina appears to be associated with activation of glutamate receptors also as the noncompetitive inhibitor of N-methyl-D-aspartate receptors dizocilpine (MK-801) partially suppressed the increase of the c-fos message. Dizocilpine Maleate 209-215 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 8298998-3 1993 Moreover, the light-induced increase of c-fos mRNA in retina appears to be associated with activation of glutamate receptors also as the noncompetitive inhibitor of N-methyl-D-aspartate receptors dizocilpine (MK-801) partially suppressed the increase of the c-fos message. Dizocilpine Maleate 209-215 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 258-263 8267594-0 1993 Evidence for enhanced expression of c-fos, c-jun, and the Ca(2+)-activated neutral protease in rat liver following carbon tetrachloride administration. Carbon Tetrachloride 115-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 8298081-0 1993 Lithium enhances 5-HT2A receptor-mediated c-fos expression in rat cerebral cortex. Lithium 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 8298081-2 1993 We have shown that the 5-HT2A/2C receptor agonist DOI (2,5-dimethoxy-4-iodophenylisopropylamine) induces the expression of c-fos in rat brain which correlates with the distribution of 5-HT2A receptors. 4-iodo-2,5-dimethoxyphenylisopropylamine 55-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 8298081-5 1993 Lithium treatment greatly enhanced levels of Fos seen after DOI, but not after 8-OH-DPAT; layer II of caudal piriform cortex, previously devoid of staining, exhibited the most marked labelling. Lithium 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 8298093-0 1993 c-fos antisense generates apomorphine and amphetamine-induced rotation. Apomorphine 26-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8298088-0 1993 Co-expression of HSP72 and c-fos in rat brain following kainic acid treatment. Kainic Acid 56-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 8298093-0 1993 c-fos antisense generates apomorphine and amphetamine-induced rotation. Amphetamine 42-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8298093-1 1993 Sodium pentobarbital anaesthetized rats were injected ipsilaterally with an antisense oligonucleotide to c-fos and contralaterally with a sense of oligonucleotide to c-fos in the striatum. Oligonucleotides 86-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 8298088-1 1993 The relationship between heat shock protein 72 (HSP72) and c-fos gene expression following systemic administration of kainic acid was investigated by combining immunocytochemistry for HSP72 with in situ hybridization for c-fos. Kainic Acid 118-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 8298093-1 1993 Sodium pentobarbital anaesthetized rats were injected ipsilaterally with an antisense oligonucleotide to c-fos and contralaterally with a sense of oligonucleotide to c-fos in the striatum. Oligonucleotides 147-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-171 8298093-4 1993 The antisense oligonucleotide also strongly inhibited the amphetamine-induced expression of c-Fos and Jun B in striatal neurones. Oligonucleotides 14-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 8298093-4 1993 The antisense oligonucleotide also strongly inhibited the amphetamine-induced expression of c-Fos and Jun B in striatal neurones. Amphetamine 58-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 8298093-5 1993 These results suggest that antisense to c-fos produces a biochemical change in the injected striatum that then, 10 h later, blocks amphetamine- and apomorphine-induced behavioural and biochemical effects. Amphetamine 131-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 8298093-5 1993 These results suggest that antisense to c-fos produces a biochemical change in the injected striatum that then, 10 h later, blocks amphetamine- and apomorphine-induced behavioural and biochemical effects. Apomorphine 148-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 8013396-8 1993 Elevated expression of c-fos and c-myc oncogenes was demonstrated 15 min after beginning treatment with BHA. Butylated Hydroxyanisole 104-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 8152604-0 1993 C-fos expression in arcuate nucleus following intracerebroventricular hypertonic saline injections. Sodium Chloride 81-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8152604-2 1993 infusions of hypertonic NaCl solutions on the induction of the protein Fos in the arcuate nucleus (Arc). Sodium Chloride 24-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 8128903-0 1993 Caffeine-induced expression of the proto-oncogene c-fos in rat striatum is increased after dopamine denervation. Caffeine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-55 8128903-0 1993 Caffeine-induced expression of the proto-oncogene c-fos in rat striatum is increased after dopamine denervation. Dopamine 91-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-55 8298088-1 1993 The relationship between heat shock protein 72 (HSP72) and c-fos gene expression following systemic administration of kainic acid was investigated by combining immunocytochemistry for HSP72 with in situ hybridization for c-fos. Kainic Acid 118-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 221-226 8298088-3 1993 Transient co-expression of c-fos and HSP72 occurred in neurons that are resistant to kainic acid, whereas prolonged co-expression was observed in vulnerable neurons. Kainic Acid 85-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 8298088-5 1993 The results demonstrate a relationship between c-fos and HSP72 gene expression and suggest that prolonged co-expression of these genes plays a role in kainic acid-induced neuronal death. Kainic Acid 151-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 8298090-0 1993 Antisense oligonucleotide to c-fos induces ipsilateral rotational behaviour to d-amphetamine. Oligonucleotides 10-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 8298090-0 1993 Antisense oligonucleotide to c-fos induces ipsilateral rotational behaviour to d-amphetamine. Dextroamphetamine 79-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 8298090-3 1993 Here we can report that direct unilateral infusion into the rat neostriatum of an antisense phosphothioate oligodeoxynucleotide to c-fos mRNA leads to the rapid induction of ipsilateral rotational behaviour after d-amphetamine administration. phosphothioate oligodeoxynucleotide 92-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-136 8298090-3 1993 Here we can report that direct unilateral infusion into the rat neostriatum of an antisense phosphothioate oligodeoxynucleotide to c-fos mRNA leads to the rapid induction of ipsilateral rotational behaviour after d-amphetamine administration. Dextroamphetamine 213-226 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-136 7504719-13 1993 Together, these results suggest that the restricted regional pattern of cocaine-induced c-fos expression is related, in part, to the basal level of dynorphin expression, and that cocaine treatment elevates dynorphin expression in striatal regions with a strong c-fos response, thereby limiting subsequent c-fos induction by cocaine. Cocaine 72-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 261-266 8254363-5 1993 c-fos mRNA and Fos protein were readily demonstrable in afferent pathways that have been implicated as mediating the neuroendocrine responses in the three stress paradigms; these include medullary catecholaminergic cell groups in response to IL-1 beta and hemorrhage, and cell groups lining the lamina terminalis in response to salt loading. Salts 328-332 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8307101-8 1993 The c-fos expression induced by a combination of NMDA (50 mg/kg) and spermidine (1.0 mumol) was unaffected by ethanol. N-Methylaspartate 49-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 8307101-8 1993 The c-fos expression induced by a combination of NMDA (50 mg/kg) and spermidine (1.0 mumol) was unaffected by ethanol. Spermidine 69-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 8307101-9 1993 Only at a high dose of ethanol (2.0 g/kg) and at minimal spermidine enhancement was NMDA-induced seizure and c-fos expression inhibited. Ethanol 23-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 7504719-0 1993 Cocaine-induced c-fos messenger RNA is inversely related to dynorphin expression in striatum. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 7504719-6 1993 A single injection of cocaine induced c-fos mRNA in striatal areas with low basal expression of dynorphin. Cocaine 22-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 7504719-11 1993 Statistical analysis of the regional patterns of basal and altered mRNA levels shows a unique inverse relationship between basal dynorphin expression and c-fos induction by cocaine. Cocaine 173-180 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 7504719-12 1993 Further evidence of this relationship is provided by the dose-dependent blockade of cocaine-induced c-fos expression by spiradoline, a dynorphin agonist. Cocaine 84-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 7504719-12 1993 Further evidence of this relationship is provided by the dose-dependent blockade of cocaine-induced c-fos expression by spiradoline, a dynorphin agonist. spiradoline 120-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 7504719-13 1993 Together, these results suggest that the restricted regional pattern of cocaine-induced c-fos expression is related, in part, to the basal level of dynorphin expression, and that cocaine treatment elevates dynorphin expression in striatal regions with a strong c-fos response, thereby limiting subsequent c-fos induction by cocaine. Cocaine 72-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 8309528-1 1993 A study was done to determine if the Fos and neurotensin immunoreactivities elicited in the rat striatal complex by the selective dopamine D2 receptor antagonist, S(-)-eticlopride hydrochloride are co-localized in the same neurons. Sulfur 163-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 8309528-1 1993 A study was done to determine if the Fos and neurotensin immunoreactivities elicited in the rat striatal complex by the selective dopamine D2 receptor antagonist, S(-)-eticlopride hydrochloride are co-localized in the same neurons. (-)-eticlopride hydrochloride 164-193 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 8309528-5 1993 Neurotensin-immunoreactive cells containing Fos nuclei represent a distinct subset, possibly the one that is dominant following administration of reserpine [Zahm (1992) Neuroscience 46, 335-350]. Reserpine 146-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 8306434-7 1993 The pattern of c-fos expression in adult animals after mechanical damage was compared with other models of focal brain injury: application of potassium to the cortical surface and devascularization. Potassium 142-151 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 8306434-8 1993 Though all models generated c-fos expression far from the lesion site, potassium application resulted in higher numbers of c-fos-positive cells, particularly in the cingulate cortex. Potassium 71-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 7504719-13 1993 Together, these results suggest that the restricted regional pattern of cocaine-induced c-fos expression is related, in part, to the basal level of dynorphin expression, and that cocaine treatment elevates dynorphin expression in striatal regions with a strong c-fos response, thereby limiting subsequent c-fos induction by cocaine. Cocaine 72-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 261-266 7504719-13 1993 Together, these results suggest that the restricted regional pattern of cocaine-induced c-fos expression is related, in part, to the basal level of dynorphin expression, and that cocaine treatment elevates dynorphin expression in striatal regions with a strong c-fos response, thereby limiting subsequent c-fos induction by cocaine. Cocaine 179-186 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 261-266 7905595-2 1993 Quantitative in situ hybridization histochemistry revealed that c-fos and zif/268 mRNAs were induced at 1 h in many limbic structures and declined 3 h after cocaine-induced convulsions. Cocaine 157-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 7504719-13 1993 Together, these results suggest that the restricted regional pattern of cocaine-induced c-fos expression is related, in part, to the basal level of dynorphin expression, and that cocaine treatment elevates dynorphin expression in striatal regions with a strong c-fos response, thereby limiting subsequent c-fos induction by cocaine. Cocaine 179-186 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 261-266 7504719-13 1993 Together, these results suggest that the restricted regional pattern of cocaine-induced c-fos expression is related, in part, to the basal level of dynorphin expression, and that cocaine treatment elevates dynorphin expression in striatal regions with a strong c-fos response, thereby limiting subsequent c-fos induction by cocaine. Cocaine 179-186 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 261-266 7504719-13 1993 Together, these results suggest that the restricted regional pattern of cocaine-induced c-fos expression is related, in part, to the basal level of dynorphin expression, and that cocaine treatment elevates dynorphin expression in striatal regions with a strong c-fos response, thereby limiting subsequent c-fos induction by cocaine. Cocaine 179-186 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 261-266 8287409-7 1993 When noradrenaline was infused, c-fos mRNA was increased fivefold after 30, threefold after 60, and 3.8-fold after 90 min. Norepinephrine 5-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 8404677-9 1993 NMA administration (regardless of the route of administration) caused an increase in LH secretion and significant cFos expression in many regions of the brain, including sites where the LHRH perikarya are concentrated. nma 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-118 8346209-2 1993 This regulatory mechanism has been proposed, in part, because the cAMP response element 2 (CRE-2) site, the key DNA regulatory element within the proenkephalin second-messenger-inducible enhancer, avidly binds AP-1 proteins, including Fos, in vitro. Cyclic AMP 66-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 235-238 8115041-2 1993 Here we report that hippocampal focal brain injury transiently induces the immediate early genes c-fos, jun-B, c-jun and krox-24 (zif-268) messenger RNA and protein and brain-derived neurotrophic factor messenger RNA in rat dentate gyrus neurons, an effect that was blocked by the N-methyl-D-aspartate receptor antagonist MK-801. Dizocilpine Maleate 322-328 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 7902768-0 1993 Autonomic areas of rat brain exhibit increased Fos-like immunoreactivity during opiate withdrawal in rats. Opiate Alkaloids 80-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 7902768-4 1993 Although some Fos-LIR was seen in these areas in control rats (either morphine-implanted, saline injected, or placebo-implanted, saline or naltrexone injected), a significantly higher number of Fos-LIR-positive cells in NTS, CVL and RVL were seen after morphine withdrawal. Sodium Chloride 90-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 7902768-4 1993 Although some Fos-LIR was seen in these areas in control rats (either morphine-implanted, saline injected, or placebo-implanted, saline or naltrexone injected), a significantly higher number of Fos-LIR-positive cells in NTS, CVL and RVL were seen after morphine withdrawal. Naltrexone 139-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 194-197 7902768-6 1993 Increased Fos-LIR was also detected in the paraventricular nucleus of the hypothalamus and the amygdala of morphine withdrawn rats. Morphine 107-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-13 8238298-5 1993 Depolarization of brain cell cultures with 50 mM K+ also caused a 100-fold increase in c-fos mRNA levels, an effect partially blocked by losartan. Losartan 137-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 8255173-7 1993 Trifluoperazine, a calcium/calmodulin inhibitor, attenuated the MTZ-induction of c-fos mRNA while potentiating the MTZ-induction of PPenk mRNA in both the hippocampus and the adrenal. Trifluoperazine 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 8255173-7 1993 Trifluoperazine, a calcium/calmodulin inhibitor, attenuated the MTZ-induction of c-fos mRNA while potentiating the MTZ-induction of PPenk mRNA in both the hippocampus and the adrenal. Pentylenetetrazole 64-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 8255173-8 1993 These results demonstrate that the MTZ induction of c-fos and Penk gene expression in the rat adrenal can be modulated by drugs acting in the CNS at NMDA receptors, in the periphery at postsynaptic cholinergic receptors and intracellularly at the calcium/calmodulin signal transduction pathway. Pentylenetetrazole 35-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 8255173-9 1993 Furthermore, we provide additional evidence that MTZ-induction of c-fos and Penk mRNAs can be dissociated by drugs acting at these sites. Pentylenetetrazole 49-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 8255178-0 1993 Acute nicotine injections induce c-fos mostly in non-dopaminergic neurons of the midbrain of the rat. Nicotine 6-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 8255178-4 1993 Acute nicotine injections resulted in prominent Fos-like immunoreactivity (-LI) in the medial terminal nucleus of the accessory optic system, the interpeduncular nucleus, and in the caudal linear subnucleus of VTA. Nicotine 6-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 8255178-6 1993 Mecamylamine abolished Fos-LI in most of the VTA neurons. Mecamylamine 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 8255178-7 1993 These results suggest that acute nicotine injections induce c-fos expression mostly in non-dopaminergic neurons of the ventral tegmental area of the rat and that nicotine induces c-fos most intensely in the interpeduncular nucleus, the superior colliculus, and several other subnuclei of the accessory optic system. Nicotine 33-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 8255178-7 1993 These results suggest that acute nicotine injections induce c-fos expression mostly in non-dopaminergic neurons of the ventral tegmental area of the rat and that nicotine induces c-fos most intensely in the interpeduncular nucleus, the superior colliculus, and several other subnuclei of the accessory optic system. Nicotine 162-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-184 8263948-8 1993 These results lend further support to the notion that Fos and related gene products mediate some of the hypertrophic actions of norepinephrine. Norepinephrine 128-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 7905609-0 1993 Haloperidol and oestrogens induce c-myc and c-fos expression in the anterior pituitary gland of the rat. Haloperidol 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 7905609-7 1993 However, in rats previously treated with oestrogens for 7 days c-myc mRNA levels increased 90 min after the injection of haloperidol and decreased to basal values after 2.5 h. c-fos mRNA levels increased sharply 30 min after haloperidol administration and also decreased to basal values 1 h later. Haloperidol 225-236 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 176-181 7902969-2 1993 A single nicotine injection resulted in a rapid and transient activation phase of nerve growth factor I-A, c-fos and jun-B at 20 min, and a later and less prominent activation of c-jun, which stayed high from 20 to 60 min. Nicotine 9-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 7902969-6 1993 When repeated nicotine injections were given, there was a refractory period of 1-2 h for c-fos, nerve growth factor I-A and jun-B induction. Nicotine 14-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 7902969-7 1993 Repeated nicotine injections at 1-h intervals prevented about 80% of c-fos, nerve growth factor I-A and jun-B mRNA induction seen after a single injection. Nicotine 9-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 7690560-6 1993 Furthermore, the expression of c-fos mRNA in VSMC was found up-regulated by 17 beta-estradiol treatment within 30 min. Estradiol 76-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 8219018-0 1993 c-Fos expression in the parabrachial nucleus after ingestion of sodium chloride in the rat. Sodium Chloride 64-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8219018-1 1993 The distribution of evoked expression of the proto-oncogene c-Fos was immunohistochemically examined in the parabrachial nucleus (PBN) of the rat after free ingestion of NaCl and some other taste solutions. Sodium Chloride 170-174 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-65 8219018-2 1993 C-Fos-like immunoreactive neurones (c-Fos neurones) were densely observed in the external lateral subnucleus (els), central lateral subnucleus (cls), and the central part of the medial subnucleus (ms). N-[(2S,3S,4R)-3,4-dihydroxy-8-oxo-8-[(4-pentylphenyl)amino]-1-{[(2S,3R,4S,5R,6R)-3,4,5-trihydroxy-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yl]oxy}octan-2-yl]hexacosanamide 110-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8219018-2 1993 C-Fos-like immunoreactive neurones (c-Fos neurones) were densely observed in the external lateral subnucleus (els), central lateral subnucleus (cls), and the central part of the medial subnucleus (ms). N-[(2S,3S,4R)-3,4-dihydroxy-8-oxo-8-[(4-pentylphenyl)amino]-1-{[(2S,3R,4S,5R,6R)-3,4,5-trihydroxy-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yl]oxy}octan-2-yl]hexacosanamide 110-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 8219018-2 1993 C-Fos-like immunoreactive neurones (c-Fos neurones) were densely observed in the external lateral subnucleus (els), central lateral subnucleus (cls), and the central part of the medial subnucleus (ms). calusterone 144-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8219018-2 1993 C-Fos-like immunoreactive neurones (c-Fos neurones) were densely observed in the external lateral subnucleus (els), central lateral subnucleus (cls), and the central part of the medial subnucleus (ms). calusterone 144-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 8219018-3 1993 The finding that the number of c-Fos neurones decreased dramatically in the ms after treatment of the tongue with amiloride or after dissection of the chorda tympani suggests that the taste information of NaCl projects mainly to the ms. Amiloride 114-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 8219018-3 1993 The finding that the number of c-Fos neurones decreased dramatically in the ms after treatment of the tongue with amiloride or after dissection of the chorda tympani suggests that the taste information of NaCl projects mainly to the ms. Sodium Chloride 205-209 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 8221069-1 1993 Subcutaneous formalin injection into the hindpaw of rats induces c-Fos expression in neurons in the ipsilateral spinal cord dorsal horn. Formaldehyde 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 8220899-5 1993 Administration of the selective CCKA receptor antagonist MK-329, but not the CCKB receptor antagonist L-365,260, prior to CCK injection, prevented oxytocin release as measured by radioimmunoassay and oxytocin neuronal activation as measured by electrophysiology and by the lack of induction of c-fos mRNA. Devazepide 57-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 294-299 8298532-2 1993 In situ hybridization was performed with a 35S-labelled 50-base oligonucleotide probe complementary to nucleotides 270-319 of rat c-fos on sections containing the pineal gland. Sulfur-35 43-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 8298532-4 1993 Stressful stimuli induced a significant increase in the expression of c-fos mRNA in the pineal gland (restraint = 144.3 +/- 14.4 cpm/mm2; hemivibrissotomy = 206.7 +/- 29.5 cpm/mm2) as compared to no restraint animals (30.6 +/- 5.1 cpm/mm2), animals displaying tonic-clonic seizures after an ip (64 mg/kg) injection of pentylenetetrazole (34.0 +/- 4.7 cpm/mm2), or competition (70.6 +/- 11.4 cpm/mm2) and RNAase-treated (52.7 +/- 9.1 cpm/mm2) controls. Pentylenetetrazole 318-336 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 8312136-5 1993 RNA was isolated and separated on denaturing gels, blotted to nylon membranes and hybridized with a 32P-labelled cDNA probe for c-fos. Phosphorus-32 100-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-133 8348687-8 1993 However, the Ca2+/cAMP response element of the c-fos gene was not sufficient to confer Ang II responsiveness to the c-fos promoter, and inhibitors of protein kinase A had no effects on Ang II-induced c-fos expression. Cyclic AMP 18-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 8348687-9 1993 On the other hand, chelating intracellular Ca2+ with BAPTA-AM inhibited Ang II-induced c-fos expression in a dose-dependent manner, suggesting that Ca2+ is required for Ang II-induced signaling. 1,2-bis(2-aminophenoxy)ethane N,N,N',N'-tetraacetic acid acetoxymethyl ester 53-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 8281324-6 1993 Oxotremorine, a non-selective muscarinic agonist, induced Fos immunoreactivity in the striatum with a large predominance in striosomes (mostly in enkephalinergic neurons), in layers 4 and 6 of the cortex, and also in the piriform cortex and septum. Oxotremorine 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 8281324-9 1993 This suggests that the induction of Fos provoked by oxotremorine does not involve dopamine, glutamate or opiates. Oxotremorine 52-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-39 8281324-10 1993 Atropine, a non-specific muscarinic antagonist, also induced Fos immunoreactivity in the striatum but with matrix predominance (mostly in substance P neurons), as well as in the cingulate cortex, and the olfactory tubercle. Atropine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 8281324-11 1993 Scopolamine, a muscarinic antagonist, induced Fos in both striosomal and matrix compartments in the striatum. Scopolamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 8229066-0 1993 Proto-oncogene c-fos induction in thiamine-deficient encephalopathy. Thiamine 34-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-20 8229066-3 1993 Induction of proto-oncogene c-fos expression, often related to seizure activity, has been detected in the brains of thiamine-deficient rats by means of Northern blot analysis and in situ hybridization. Thiamine 116-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-33 8366353-9 1993 Administration of imipramine alone under basal conditions produced a robust induction of Fos-LI in the central nucleus of the amygdala and in the dorsal lateral subdivision of the bed nucleus of the stria terminalis. Imipramine 18-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 8255396-3 1993 Sexual behavior significantly increased the percentage of OT neurons in the hypothalamic paraventricular nucleus (PVN) expressing FOS in rats treated with estradiol and progesterone, compared to hormone-treated, nonmated controls. Estradiol 155-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-133 8255396-3 1993 Sexual behavior significantly increased the percentage of OT neurons in the hypothalamic paraventricular nucleus (PVN) expressing FOS in rats treated with estradiol and progesterone, compared to hormone-treated, nonmated controls. Progesterone 169-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-133 8233100-1 1993 Experiments were done in conscious rats to investigate the effect of intravenous infusion of hypertonic saline on the induction of the protein Fos, in brainstem neurons. Sodium Chloride 104-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-146 8233100-2 1993 Neurons containing Fos-like immunoreactivity were observed in the caudal nucleus of the solitary tract (NTS), the caudal and rostral ventrolateral medulla, and parabrachial nucleus after an infusion of solutions containing 1.4 M NaCl. Sodium Chloride 229-233 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 8102797-0 1993 Striatal Fos expression is indicative of dopamine D1/D2 synergism and receptor supersensitivity. Dopamine 41-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 8102797-4 1993 Using immunocytochemical methods in rats treated with directly acting selective dopamine agonists, we have determined that dopamine-mediated expression of Fos and Fos-like antigens in the striatum normally requires concomitant stimulation of D1 and D2 receptors. Dopamine 80-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-158 8102797-4 1993 Using immunocytochemical methods in rats treated with directly acting selective dopamine agonists, we have determined that dopamine-mediated expression of Fos and Fos-like antigens in the striatum normally requires concomitant stimulation of D1 and D2 receptors. Dopamine 80-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 8102797-4 1993 Using immunocytochemical methods in rats treated with directly acting selective dopamine agonists, we have determined that dopamine-mediated expression of Fos and Fos-like antigens in the striatum normally requires concomitant stimulation of D1 and D2 receptors. Dopamine 123-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-158 8102797-4 1993 Using immunocytochemical methods in rats treated with directly acting selective dopamine agonists, we have determined that dopamine-mediated expression of Fos and Fos-like antigens in the striatum normally requires concomitant stimulation of D1 and D2 receptors. Dopamine 123-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 8102797-5 1993 Separate administration of a high dose of a selective D1 (SKF 38393; 20 mg/kg) or D2 (quinpirole; 3 mg/kg) agonist induced Fos-like immunoreactivity in few neurons, whereas combined administration of the D1 and D2 agonists produced patches of intensely stained immunoreactive nuclei in the caudate-putamen. Quinpirole 86-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-126 8102797-7 1993 These data demonstrate that dopamine-mediated Fos expression in the striatum is indicative of the state of D1/D2 synergism and receptor supersensitivity. Dopamine 28-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 8356069-3 1993 Also detected in salt-loaded animals was a prominent induction of the immediate-early gene product Fos in magnocellular neurosecretory cell groups and in several brain regions that are known to provide major projections to the endocrine hypothalamus. Salts 17-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 8368314-0 1993 Decreased expression of protooncogenes c-fos, c-myc, and c-jun following polyamine depletion in IEC-6 cells. Polyamines 73-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 8368314-2 1993 This study tests the hypothesis that the protooncogenes c-fos, c-myc, c-jun, and junB are involved in the mechanism by which polyamines modulate mucosal growth. Polyamines 125-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 8368314-9 1993 c-fos mRNA in quiescent cells after 24 h serum deprivation was significantly stimulated by 5% dFBS, although a steady-state level of c-fos mRNA was undetectable in control cells. dfbs 94-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8281310-0 1993 Transection of corticostriatal afferents reduces amphetamine- and apomorphine-induced striatal Fos expression and turning behaviour in unilaterally 6-hydroxydopamine-lesioned rats. Amphetamine 49-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 8281310-0 1993 Transection of corticostriatal afferents reduces amphetamine- and apomorphine-induced striatal Fos expression and turning behaviour in unilaterally 6-hydroxydopamine-lesioned rats. Apomorphine 66-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 8281310-8 1993 The frontocortical transection reduced both apomorphine- and amphetamine-induced Fos expression by 33-66% within the ipsilateral caudate-putamen. Apomorphine 44-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-84 8281310-8 1993 The frontocortical transection reduced both apomorphine- and amphetamine-induced Fos expression by 33-66% within the ipsilateral caudate-putamen. Amphetamine 61-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-84 8102782-0 1993 Metrazole induces the sequential activation of c-fos, proenkephalin, and tyrosine hydroxylase gene expression in the rat adrenal gland: modulation by glucocorticoid and adrenocorticotropic hormone. Pentylenetetrazole 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 8102782-3 1993 We investigated the relationships between the activation of the c-fos gene and the activation of the Penk and TH genes in both rat hippocampus and adrenal using a commonly used model, metrazole (MTZ)-induced convulsions. Pentylenetetrazole 184-193 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 8102782-3 1993 We investigated the relationships between the activation of the c-fos gene and the activation of the Penk and TH genes in both rat hippocampus and adrenal using a commonly used model, metrazole (MTZ)-induced convulsions. Pentylenetetrazole 195-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 8102782-4 1993 The administration of MTZ produced a sequential elevation in c-fos, preproenkephalin (PPenk), and TH mRNAs. Pentylenetetrazole 22-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 8102782-5 1993 One hour after MTZ administration, c-fos mRNA was increased about 10-fold in rat hippocampus and about 5-fold in rat adrenal, without a significant change in spinal cord levels. Pentylenetetrazole 15-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 8102782-6 1993 Immunocytochemistry revealed that Fos-like immunoreactivity was greatly increased in both hippocampus and adrenal medulla at 3 hr after MTZ administration. Pentylenetetrazole 136-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 8102782-8 1993 The effects of MTZ on c-fos, PPenk, and TH mRNAs were dose dependent in both adrenal and hippocampus. Pentylenetetrazole 15-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 8102782-11 1993 ACTH treatment completely blocked the MTZ induction of adrenal c-fos mRNA and the subsequent induction of Fos-like immunoreactivity, whereas MTZ increased PPenk and TH mRNAs nearly 3-fold. Pentylenetetrazole 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 8102782-12 1993 Thus, in hypox rats MTZ can increase adrenal c-fos and TH mRNA levels without a corresponding increase in PPenk mRNA, whereas in ACTH-treated rats PPenk and TH mRNA levels in adrenal can be increased by MTZ without a preceding increase in c-fos mRNA. Pentylenetetrazole 20-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 8102782-12 1993 Thus, in hypox rats MTZ can increase adrenal c-fos and TH mRNA levels without a corresponding increase in PPenk mRNA, whereas in ACTH-treated rats PPenk and TH mRNA levels in adrenal can be increased by MTZ without a preceding increase in c-fos mRNA. Pentylenetetrazole 20-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 239-244 8102782-12 1993 Thus, in hypox rats MTZ can increase adrenal c-fos and TH mRNA levels without a corresponding increase in PPenk mRNA, whereas in ACTH-treated rats PPenk and TH mRNA levels in adrenal can be increased by MTZ without a preceding increase in c-fos mRNA. Pentylenetetrazole 203-206 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 239-244 8102782-13 1993 The MTZ induction of c-fos appears neither sufficient nor always necessary for the subsequent MTZ induction of Penk and TH gene expression. Pentylenetetrazole 4-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 8404677-4 1993 Treatment with MK-801 (0.3 mg/kg, sc) at 1130 h blocked both the LH and PRL surges and cFos expression in LHRH neurons. Dizocilpine Maleate 15-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-91 8404677-6 1993 We then determined whether NMA treatment could restore LH secretion and cFos expression in LHRH neurons in animals whose endogenous proestrous LH surges were blocked with pentobarbital. nma 27-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-76 8221117-0 1993 L-dopa stimulates c-fos expression in dopamine denervated striatum by combined activation of D-1 and D-2 receptors. Levodopa 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 8221117-0 1993 L-dopa stimulates c-fos expression in dopamine denervated striatum by combined activation of D-1 and D-2 receptors. Dopamine 38-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 8221117-1 1993 Administration of L-dopa to unilaterally 6-hydroxydopamine-lesioned rats, activates the early gene c-fos in the lesioned caudate-putamen. Levodopa 18-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 8221117-1 1993 Administration of L-dopa to unilaterally 6-hydroxydopamine-lesioned rats, activates the early gene c-fos in the lesioned caudate-putamen. Oxidopamine 41-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 8221117-2 1993 D-1 receptor blockade by SCH 23390, prevented L-dopa-induced Fos-like immunoreactivity in the whole caudate-putamen, while D-2 receptor blockade by raclopride reduced Fos-like immunoreactivity only in the dorso-lateral portion. Levodopa 46-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 8221117-2 1993 D-1 receptor blockade by SCH 23390, prevented L-dopa-induced Fos-like immunoreactivity in the whole caudate-putamen, while D-2 receptor blockade by raclopride reduced Fos-like immunoreactivity only in the dorso-lateral portion. Raclopride 148-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 167-170 8221117-3 1993 The results suggest that L-dopa induces c-fos primarily through an activation of D-1 receptors, while D-2 receptor stimulation plays a facilitatory influence on D-1-mediated c-fos expression. Levodopa 25-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 8255173-0 1993 Metrazole induction of c-fos and proenkephalin gene expression in the rat adrenal and hippocampus: pharmacological characterization. Pentylenetetrazole 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 8255173-1 1993 We have previously reported that the administration of metrazole (MTZ) produces a sequential, dose-dependent induction of c-fos and proenkephalin (Penk) gene expression in the rat hippocampus and adrenal. Pentylenetetrazole 55-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 8255173-1 1993 We have previously reported that the administration of metrazole (MTZ) produces a sequential, dose-dependent induction of c-fos and proenkephalin (Penk) gene expression in the rat hippocampus and adrenal. Pentylenetetrazole 66-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 8255173-4 1993 Treatment with LY274614, a competitive NMDA-receptor antagonist, blocked MTZ-induced convulsions and the MTZ-induction of c-fos and PPenk mRNAs in the hippocampus, and PPenk mRNA in the adrenal. LY 274614 15-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 8255173-4 1993 Treatment with LY274614, a competitive NMDA-receptor antagonist, blocked MTZ-induced convulsions and the MTZ-induction of c-fos and PPenk mRNAs in the hippocampus, and PPenk mRNA in the adrenal. Pentylenetetrazole 105-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 8255173-5 1993 However, in the adrenal the MTZ-induction of c-fos was only partially inhibited by LY274614. Pentylenetetrazole 28-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 8255173-6 1993 A combination of peripheral acting cholinergic antagonists (chlorisondamine plus methylatropine) prevented the MTZ-induction of adrenal c-fos and PPenk mRNA without significant alterations in the MTZ-induction of hippocampal c-fos mRNA or convulsions. Chlorisondamine 60-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 8255173-6 1993 A combination of peripheral acting cholinergic antagonists (chlorisondamine plus methylatropine) prevented the MTZ-induction of adrenal c-fos and PPenk mRNA without significant alterations in the MTZ-induction of hippocampal c-fos mRNA or convulsions. methylatropine 81-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 8255173-6 1993 A combination of peripheral acting cholinergic antagonists (chlorisondamine plus methylatropine) prevented the MTZ-induction of adrenal c-fos and PPenk mRNA without significant alterations in the MTZ-induction of hippocampal c-fos mRNA or convulsions. Pentylenetetrazole 111-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 8255173-6 1993 A combination of peripheral acting cholinergic antagonists (chlorisondamine plus methylatropine) prevented the MTZ-induction of adrenal c-fos and PPenk mRNA without significant alterations in the MTZ-induction of hippocampal c-fos mRNA or convulsions. Pentylenetetrazole 111-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 225-230 8404612-0 1993 Thyrotropin-releasing hormone stimulates c-jun and c-fos messenger ribonucleic acid levels: implications for calcium mobilization and protein kinase-C activation. Calcium 109-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 8404612-8 1993 Our data indicate that calcium mobilization and protein kinase activation represent distinct components of the signaling events initiated by TRH resulting in increased c-jun and c-fos mRNA levels. Calcium 23-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 178-183 8263948-0 1993 Expression of c-fos and related genes in the rat heart in response to norepinephrine. Norepinephrine 70-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 8263948-1 1993 We have investigated the cellular and regional localization of Fos-like immunoreactivity (FLI) in the rat heart in response to the hypertrophic hormone norepinephrine (NE). Norepinephrine 152-166 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 8263948-4 1993 The response was transient with maximal immunoreactivity observed 2-3h following treatment, falling to near basal levels in most regions of the heart after 6 h. Although all chambers of the heart contributed to the response, greatest Fos-like immunoreactivity was observed in the left atrium and left ventricle, with intermediate levels found in the septum and right ventricle, and lowest levels in the right atrium. Tritium 68-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 234-237 8292488-0 1993 Cycloheximide prevents kainate-induced neuronal death and c-fos expression in adult rat brain. Cycloheximide 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 8292488-5 1993 Pretreatment with cycloheximide did not affect the transient induction of c-fos observed in numerous structures, but significantly reduced the prolonged expression of c-fos mRNA in kainate-vulnerable regions. Cycloheximide 18-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 167-172 8292488-9 1993 These results not only demonstrate that kainate-induced neurotoxicity and the prolonged expression of c-fos are both prevented by cycloheximide, but also strengthen idea that prolonged c-fos expression is a marker of neuronal death. Kainic Acid 40-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 185-190 8292488-9 1993 These results not only demonstrate that kainate-induced neurotoxicity and the prolonged expression of c-fos are both prevented by cycloheximide, but also strengthen idea that prolonged c-fos expression is a marker of neuronal death. Cycloheximide 130-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 8346209-3 1993 However, we observe a dissociation in the time course of activation of c-fos and proenkephalin mRNA in rat striatum after administration of the dopamine D2 receptor antagonist haloperidol. Haloperidol 176-187 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 8346209-5 1993 Even though our striatal nuclear extracts had substantial basal and haloperidol-inducible AP-1-binding activities that contained Fos, we could not detect Fos in complexes bound to the CRE-2 element. Haloperidol 68-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-132 8343151-4 1993 Therefore we investigated the effect of hypertonic NaCl and sucrose solutions on the transcriptional activation of the immediate-early genes (IEGs) egr-1 and c-fos in isolated ventricular adult rat cardiomyocytes. Sucrose 60-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 19912939-8 1993 There is a different time course of expression for the Fos and Fra family of genes after in vivo NMDA. N-Methylaspartate 97-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 19912939-0 1993 Intrahippocampal NMDA Administration Alters Fos, Fos-Related Antigens, and Opioid Peptide Immunoreactivity and mRNA in Rats. N-Methylaspartate 17-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 19912939-0 1993 Intrahippocampal NMDA Administration Alters Fos, Fos-Related Antigens, and Opioid Peptide Immunoreactivity and mRNA in Rats. N-Methylaspartate 17-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 19912939-2 1993 Fos-ir and Fra-ir were observed in the dentate granule cells and nonpyramidal cells within the hippocampal formation at 1 h, and in all hippocampal cells and fields, 3 h following injection of NMDA into the ventral hippocampus. N-Methylaspartate 193-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 8345107-10 1993 Following KCl or BIC, a strong induction was seen for c-FOS, JUN B, and KROX-24, whereas c-JUN, JUN D, and FOS B showed only a moderate increase compared to basal levels. Potassium Chloride 10-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 8233031-3 1993 Both lithium chloride and osmotic pressure enhanced c-fos expression in the nuclei of the solitary tract, the paraventricular nuclei and supraoptic nuclei of the hypothalamus, and in the amygdala. Lithium Chloride 5-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 8358602-15 1993 injection of saline-induced expression of c-fos mRNA in the piriform cortex, neocortex, cingulate cortex and the amygdala was much less than that seen after i.c.v.-administered CRF as evident in the intensity of the signals. Sodium Chloride 13-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 8345107-10 1993 Following KCl or BIC, a strong induction was seen for c-FOS, JUN B, and KROX-24, whereas c-JUN, JUN D, and FOS B showed only a moderate increase compared to basal levels. Bicuculline 17-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 8345107-16 1993 The present data demonstrate that CREB, JUN, FOS, and KROX transcription factors exhibit a layer-specific expression in the cerebral cortex with only slight area-specific differences both in untreated rats and following stimulation with KCl and BIC. Potassium Chloride 237-240 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 8233031-0 1993 Expression of c-fos in brain subcortical structures in response to nauseant lithium chloride and osmotic pressure in rats. Lithium Chloride 76-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 8345107-16 1993 The present data demonstrate that CREB, JUN, FOS, and KROX transcription factors exhibit a layer-specific expression in the cerebral cortex with only slight area-specific differences both in untreated rats and following stimulation with KCl and BIC. Bicuculline 245-248 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 8342693-3 1993 These findings support the hypothesis that CCK-8 induced c-fos expression is mediated by CCK-A receptors. Sincalide 43-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 8103187-1 1993 In this paper, we tested whether physiological activators of the cAMP second messenger pathway in primary cultures of neurons from rat cerebral cortex directly induce c-fos and other immediate early gene (IEG) transcription factors. Cyclic AMP 65-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 167-172 8319588-0 1993 The antiestrogen tamoxifen induces c-fos and jun-B, but not c-jun or jun-D, protooncogenes in the rat uterus. Tamoxifen 17-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 8319588-4 1993 The purpose of the present study was to investigate the effect of tamoxifen on activation of the c-fos, c-jun, jun-B and jun-D protooncogenes in rat uteri in vivo. Tamoxifen 66-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 8319588-6 1993 Northern blotting analysis of total uterine RNA revealed that treatment of adult, ovariectomized rats with tamoxifen elevated levels of c-fos mRNA 2.4-, 5.3- and 6.2-fold over expression in untreated rats by 6, 12 and 24 h post-tamoxifen injection, respectively. Tamoxifen 107-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 8319588-6 1993 Northern blotting analysis of total uterine RNA revealed that treatment of adult, ovariectomized rats with tamoxifen elevated levels of c-fos mRNA 2.4-, 5.3- and 6.2-fold over expression in untreated rats by 6, 12 and 24 h post-tamoxifen injection, respectively. Tamoxifen 228-237 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 8319588-7 1993 Tamoxifen induction of c-fos was still apparent by 48 h post injection. Tamoxifen 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 8377936-0 1993 Stimulation of dopamine transmission in the dorsal caudate nucleus by pargyline as demonstrated by dopamine and acetylcholine microdialysis and Fos immunohistochemistry. Dopamine 15-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-147 8377936-1 1993 The effect of pargyline on dopamine neurotransmission was investigated by trans-striatal microdialysis combined with Fos immunohistochemistry. Pargyline 14-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 8377936-3 1993 Administration of pargyline resulted in the appearance of Fos-positive nuclei distributed along a gradient around the dialysis probe. Pargyline 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 8377936-5 1993 Similarly, lack of calcium in the perfusion medium abolished the effect of pargyline on dopamine and acetylcholine output and on Fos formation. Calcium 19-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-132 8377936-6 1993 In rats not implanted with dialysis probes pargyline administration resulted in only rare Fos-positive nuclei in the dorsal caudate. Pargyline 43-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 8337705-0 1993 Trimethyltin-induced c-fos expression: adolescent vs neonatal rat hippocampus. trimethyltin 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 8414190-5 1993 The expression of c-fos, c-jun, jun-B, and NGFI-A mRNA are rapidly induced in cultured astrocytes after treatment with phorbol ester, epidermal growth factor, and basic fibroblast growth factor. Phorbol Esters 119-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 8353932-0 1993 Age- and dose-related NMDA induction of Fos-like immunoreactivity and c-fos mRNA in the arcuate nucleus of immature female rats. N-Methylaspartate 22-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 8353932-0 1993 Age- and dose-related NMDA induction of Fos-like immunoreactivity and c-fos mRNA in the arcuate nucleus of immature female rats. N-Methylaspartate 22-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 8353932-2 1993 Peripheral injection of prepubertal rats with NMDA induces maximal secretion of LH within 8 min as well as the expression of the proto-oncogene, c-fos, within the mediobasal hypothalamus (MBH). N-Methylaspartate 46-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-150 8353932-5 1993 Our results indicate that NMDA-induced c-fos mRNA and protein are maximal by 60 and 120 min, respectively. N-Methylaspartate 26-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 8353932-6 1993 Both c-fos mRNA and protein attain peak levels using NMDA doses between 20 and 40 mg/kg. N-Methylaspartate 53-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 5-10 8353932-8 1993 The data presented here indicate that the immunohistochemical localization of c-fos gene expression, in conjunction with in situ hybridization, is a useful technique for mapping NMDA-sensitive pathways and may provide anatomical and physiological evidence that better defines the glutamatergic control of sexual maturation. N-Methylaspartate 178-182 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 8356885-0 1993 Impairment of Fos protein formation in the rat infarct borderzone by MK-801, but not by NBQX. Dizocilpine Maleate 69-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 8356885-3 1993 The saline treated rats showed presence of Fos protein in nerve cell nuclei throughout the cortical and striatal infarct borderzone, but no staining in the infarct core or contralateral hemisphere. Sodium Chloride 4-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 8356885-4 1993 MK-801 almost totally abolished this expression of Fos protein whereas NBQX had no significant effect on Fos protein expression. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 8326829-1 1993 SKF 38,393, a dopamine receptor agonist which stimulates the D1 receptor, has been shown to activate c-fos to a detectable level in rats only when the striatum has been sensitized by a previous 6-OHDA lesion of the nigrostriatal tract. skf 38 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 8514261-5 1993 On the contrary, the hyperplasia induced by cyproterone acetate and nafenopin, which is characterized by a lack of increase in the expression of c-fos and c-jun, was also not associated with an increased c-myc expression. Nafenopin 68-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-150 8501519-5 1993 MK-801 additionally prevents the transcriptional activation of several immediate-early genes (IEGs) (e.g., c-fos) and their cognate third nuclear messenger (i.e., c-Fos) expression present in the hemisphere ipsilateral to the lesion. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 8501519-5 1993 MK-801 additionally prevents the transcriptional activation of several immediate-early genes (IEGs) (e.g., c-fos) and their cognate third nuclear messenger (i.e., c-Fos) expression present in the hemisphere ipsilateral to the lesion. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-168 19912927-0 1993 Regional Effects of NMDA Receptor Antagonists on the Induction of Striatal c-fos by Fenfluramine. Fenfluramine 84-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 19912927-4 1993 Systemic (IP) administration of the above receptor antagonists 30 min prior to fenfluramine exposure resulted in the blockade of c-fos immunoreactivity. Fenfluramine 79-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 19912927-6 1993 In addition, pretreatment with the D(1) receptor antagonist SCH 23390 (0.5 mg/kg) also partially suppressed the induction of c-fos by fenfluramine. SCH 23390 60-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 19912927-6 1993 In addition, pretreatment with the D(1) receptor antagonist SCH 23390 (0.5 mg/kg) also partially suppressed the induction of c-fos by fenfluramine. Fenfluramine 134-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 19912927-7 1993 These findings suggest the participation of NMDA as well as dopamine-sensitive receptors for the induction of striatal c-fos by an indirect serotonergic (5-HT) agonist. N-Methylaspartate 44-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 8332263-1 1993 Immunocytochemistry was used to assess the distribution of neurons within the spinal trigeminal nucleus that expressed the protein product of the proto-oncogene c-fos after thermal stimulation of the cornea in barbiturate-anesthetized rats. barbituric acid 210-221 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-166 8332263-10 1993 The appearance of Fos-like immunoreactivity within caudal portions of the nucleus was increased only by noxious intensities of stimulation and was further enhanced in animals with low levels of corticosterone. Corticosterone 194-208 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 8347819-1 1993 The laminar distribution of spinal cord neurones expressing immunoreactivity to the Fos protein was evaluated in the rat following chemical, thermal or mechanical noxious stimulation of the skin for 2 h. After stimulation by 20% or 5% formalin, Fos-immunoreactive neurones prevailed in lamina I where they accounted for 64% and 59%, respectively, of the entire population of Fos-immunoreactive spinal cells. Formaldehyde 235-243 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 8503903-0 1993 Differential modulation of fos and jun gene expression by 1,25-dihydroxyvitamin D3. Calcitriol 58-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 8503903-6 1993 Thus 1,25-(OH)2D3 dramatically alters the ratio of jun/fos mRNA produced during the mitogenic response and may thus modulate the response to mitogens. Calcitriol 5-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 8319761-0 1993 C-fos antisense in the nucleus accumbens blocks the locomotor stimulant action of cocaine. Cocaine 82-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8319761-1 1993 Systemic cocaine induces c-fos expression in rat striatum. Cocaine 9-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 8319761-4 1993 Here, we report that bilateral administration of an antisense oligo against c-fos in the nucleus accumbens blocks cocaine induced locomotor stimulation, without affecting spontaneous exploratory activity. Cocaine 114-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 8319761-6 1993 This finding suggests a role for c-fos in mediating psychostimulant effects of cocaine. Cocaine 79-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 8361643-4 1993 We found that deep noxious stimulation led to Fos-positive cells predominantly in the ventrolateral PAG and superficial noxious stimulation led to Fos-positive cells predominantly in the lateral PAG. pag 100-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 8361643-4 1993 We found that deep noxious stimulation led to Fos-positive cells predominantly in the ventrolateral PAG and superficial noxious stimulation led to Fos-positive cells predominantly in the lateral PAG. pag 195-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-150 8098756-10 1993 In support of a regional action of MK-801, MK-801 induced c-fos-like immunoreactivity in the cerebral cortex, but not in the nucleus accumbens or striatum. Dizocilpine Maleate 35-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 8098756-10 1993 In support of a regional action of MK-801, MK-801 induced c-fos-like immunoreactivity in the cerebral cortex, but not in the nucleus accumbens or striatum. Dizocilpine Maleate 43-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 8098756-11 1993 The action of MK-801 to increase c-fos-like immunoreactivity in cerebral cortex was reduced, but not blocked, by SCH-23390. Dizocilpine Maleate 14-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 8098756-12 1993 Additionally, MK-801 reduced, but did not eliminate, D1-dopamine agonist induced c-fos-like immunoreactivity in striatum. Dizocilpine Maleate 14-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 8098756-12 1993 Additionally, MK-801 reduced, but did not eliminate, D1-dopamine agonist induced c-fos-like immunoreactivity in striatum. Dopamine 56-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 8099817-1 1993 The impact of cocaine self-administration on the expression of striatal preprodynorphin and c-fos mRNA was examined with in situ hybridization histochemistry. Cocaine 14-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 8099817-5 1993 Furthermore, these data indicate that the regulation of preprodynorphin is dissociable from the expression of c-fos in rats exposed to repeated doses of cocaine. Cocaine 153-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 8502228-0 1993 Tamoxifen stimulates expression of the c-fos proto-oncogene in rodent uterus. Tamoxifen 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 8502228-4 1993 Tamoxifen increases the 2.2-kilobase c-fos transcript approximately 20-fold in 6 hr. Tamoxifen 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 8502228-6 1993 c-fos induction is observed at doses of 0.1-10 mg/kg tamoxifen. Tamoxifen 53-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8502228-7 1993 The nonsteroidal antiestrogens nafoxidine, Cl-628, and 4-hydroxytamoxifen also induce c-fos expression. Nafoxidine 31-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 8502228-7 1993 The nonsteroidal antiestrogens nafoxidine, Cl-628, and 4-hydroxytamoxifen also induce c-fos expression. cl-628 43-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 8502228-7 1993 The nonsteroidal antiestrogens nafoxidine, Cl-628, and 4-hydroxytamoxifen also induce c-fos expression. hydroxytamoxifen 55-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 8502228-8 1993 The induction of c-fos by both estradiol and tamoxifen is blocked by the progestin medroxyprogesterone acetate. Estradiol 31-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 8502228-8 1993 The induction of c-fos by both estradiol and tamoxifen is blocked by the progestin medroxyprogesterone acetate. Tamoxifen 45-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 8502228-8 1993 The induction of c-fos by both estradiol and tamoxifen is blocked by the progestin medroxyprogesterone acetate. Medroxyprogesterone Acetate 83-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 8100045-9 1993 Both medetomidine doses administered 12 min before formalin strongly suppressed the expression of c-FOS in the spinal dorsal horn; the suppression was stronger in the deep (III-VI) than in the superficial (I and II) laminae of the dorsal horn (76% and 86% for 100 micrograms/kg dose vs 97% and 99% for 300 micrograms/kg dose, respectively). Medetomidine 5-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 8100045-11 1993 In the parabrachial nucleus, both medetomidine doses also produced a significant suppression of c-FOS expression (68%). Medetomidine 34-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 8100045-15 1993 However, in the medial thalamus, atipamezole produced a dramatic increase of formalin-induced c-FOS expression when compared with formalin injection alone. atipamezole 33-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 8100045-15 1993 However, in the medial thalamus, atipamezole produced a dramatic increase of formalin-induced c-FOS expression when compared with formalin injection alone. Formaldehyde 77-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 8100045-19 1993 Compared with the previously reported effects of behaviorally equipotent doses of morphine, the suppression of c-FOS expression in the spinal cord was stronger following medetomidine than that following morphine. Morphine 82-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 8100045-19 1993 Compared with the previously reported effects of behaviorally equipotent doses of morphine, the suppression of c-FOS expression in the spinal cord was stronger following medetomidine than that following morphine. Medetomidine 170-182 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 8100045-19 1993 Compared with the previously reported effects of behaviorally equipotent doses of morphine, the suppression of c-FOS expression in the spinal cord was stronger following medetomidine than that following morphine. Morphine 203-211 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 8511714-0 1993 Expression of c-fos protein in the experimental epilepsy induced by pilocarpine. Pilocarpine 68-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 8511714-1 1993 The expression of the c-fos proto-oncogene, as estimated by immunohistochemistry of the FOS nuclear protein, was studied in both focal and generalized seizures induced in rats by systemic administration of pilocarpine. Pilocarpine 206-217 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 8484751-4 1993 We reported that in rat thymocytes, undergoing cell proliferation upon stimulation with Concanavalin A or cell death following dexamethasone treatment, an induction of c-fos and c-jun occurs with different but partially overlapping kinetics (Grassilli et al., BBRC, 1992, 188, 1261-1266). Dexamethasone 127-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-173 8508307-1 1993 Northern blot analysis was used to study the effects of acrylamide, a potent neurotoxin, on the induction of c-fos and c-jun mRNA in rat brain. Acrylamide 56-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 8508307-4 1993 Acute administration of acrylamide caused a statistically significant increase in the expression of c-fos (approx. Acrylamide 24-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 8508307-7 1993 By contrast, the level of c-fos mRNA in chronic acrylamide treatment was not altered significantly, but the expression of c-jun mRNA was increased almost 100% as compared to control. Acrylamide 48-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 8386370-1 1993 To investigate the mechanisms of asbestos-induced carcinogenesis, expression of c-fos and c-jun protooncogenes was examined in rat pleural mesothelial cells and hamster tracheal epithelial cells after exposure to crocidolite or chrysotile asbestos. Asbestos 33-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 8099723-0 1993 Glutamate induces the growth factors NGF, bFGF, the receptor FGF-R1 and c-fos mRNA expression in rat astrocyte culture. Glutamic Acid 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 8099723-2 1993 Incubation with glutamate (100 microM, 15 min) induced nerve growth factor (NGF), basic fibroblast growth factor (bFGF), FGF receptor (FGF-R1) and proto-oncogene c-fos gene expression in a time dependent manner. Glutamic Acid 16-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-167 8386370-3 1993 The induction of c-fos and c-jun mRNA by asbestos in mesothelial cells is dose-dependent and is most pronounced with crocidolite, the type of asbestos most pathogenic in the causation of pleural mesothelioma. Asbestos, Crocidolite 117-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 8481796-3 1993 The results indicate that the number of Fos-like immunoreactive neurons (1) is slightly decreased in ketamine-anesthetized non-stimulated arthritic rats as compared to the non-anesthetized non-stimulated ones, (2) is significantly higher in both stimulated normal and arthritic animals as compared to non-stimulated animals, particularly in laminae I, II, V and VI of L3 and L4, and (3) is significantly increased in stimulated arthritic as compared to stimulated normal rats, in all laminae of lumbar spinal segments. Ketamine 101-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 8515876-0 1993 On the expression of Fos-like protein in the subthalamic nucleus and basal ganglia output systems following kainic acid injections into the rodent striatum. Kainic Acid 108-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 8515876-1 1993 By immunohistochemistry, Fos-like protein induced in response to intrastriatal kainic acid injections was explored in the basal ganglia and the ventrolateral thalamic nucleus of the rat. Kainic Acid 79-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 8515876-2 1993 In the globus pallidus, entopeduncular nucleus, substantia nigra pars reticulata, subthalamic nucleus and ventrolateral thalamic nucleus, Fos was expressed in neuronal cell nuclei showing the maximal number at 4 h and disappeared within 48 h. The substantia nigra pars compacta neurons lacked prominent Fos induction, suggesting that a different mechanism may operate for changes of dopamine systems. Dopamine 383-391 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-141 8491280-0 1993 Levodopa induction of Fos immunoreactivity in rat brain following partial and complete lesions of the substantia nigra. Levodopa 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-25 8491280-3 1993 We have investigated the potential effects of L-DOPA in early PD by analyzing its influence on the activity of the immediate early gene Fos in the partially lesioned rat. Levodopa 46-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-139 8491280-4 1993 Fos has been used to examine neuronal response to a variety of stimuli in vivo, and L-DOPA is known to increase Fos-like immunoreactivity (FosLI) in striatal neurons after complete lesions of the SN. Levodopa 84-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 8491280-10 1993 Therefore L-DOPA induces Fos in the striatum after partial as well as complete SN lesions. Levodopa 10-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 8491280-11 1993 This result suggests that L-DOPA-induced Fos expression may occur in the early stages of PD and may not require dopamine receptor upregulation, which is believed to develop only in completely lesioned rats and in later stages of PD. Levodopa 26-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 8491280-13 1993 Since Fos is known to regulate gene transcription, potential alterations in its activity may contribute to the complications associated with L-DOPA therapy in PD. Levodopa 141-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 6-9 8386370-6 1993 The persistent induction of AP-1 transcription factors by asbestos suggests a model of asbestos-induced carcinogenesis involving chronic stimulation of cell proliferation through activation of the early response gene pathway that includes c-jun and/or c-fos. Asbestos 58-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 252-257 8386370-6 1993 The persistent induction of AP-1 transcription factors by asbestos suggests a model of asbestos-induced carcinogenesis involving chronic stimulation of cell proliferation through activation of the early response gene pathway that includes c-jun and/or c-fos. Asbestos 87-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 252-257 8479154-2 1993 EXPERIMENTAL DESIGN: We have studied the time course of the induction and the effects of cycloheximide treatment on the expression of c-fos, c-jun and the heat-shock gene HSP 70 in ischemic-reperfused livers; extracts of these livers have also been examined for the binding to a synthetic oligonucleotide containing the heat-shock consensus sequence (HSE) in order to reveal the possible presence of an active heat-shock factor (HSF) in ischemic-reperfused tissue. Cycloheximide 89-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-139 8328305-2 1993 Injury produced by infusion of 5 microliters of saline into the hippocampus produced a time-dependent expression of Fos, Jun and Krox 24, but not Fos B, Krox 20 or in non-nerve cells around the wound margin, in cells lining the lateral and third ventricles and in cells in the pial surfaces of the brain. Sodium Chloride 48-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 8098138-0 1993 Blockade of muscarinic receptors potentiates D1 dependent turning behavior and c-fos expression in 6-hydroxydopamine-lesioned rats but does not influence D2 mediated responses. Oxidopamine 99-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 8098138-2 1993 Studies on the expression of the early gene c-fos as reflected by the immunohistochemical demonstration of the Fos protein, show that administration of scopolamine (5 mg/kg, i.p.) Scopolamine 152-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 8098138-2 1993 Studies on the expression of the early gene c-fos as reflected by the immunohistochemical demonstration of the Fos protein, show that administration of scopolamine (5 mg/kg, i.p.) Scopolamine 152-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 8098138-3 1993 potentiates c-fos expression elicited by SKF 38393 (1.5 mg/kg, s.c.) in the caudate-putamen of the lesioned side. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 41-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 8492912-1 1993 Immunocytochemistry has been used to monitor the expression of the immediate-early gene c-fos in rat brain following administration of the serotonin2/1C receptor agonist 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane. 4-iodo-2,5-dimethoxyphenylisopropylamine 170-215 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 8100600-8 1993 Fos was visualized with nickel-intensified diaminobenzidine (Ni-DAB) in the first sequence while TH, PNMT, OT or VP were visualized with DAB alone, resulting in readily distinguishable black and amber reaction products, respectively. Nickel 24-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 8100600-8 1993 Fos was visualized with nickel-intensified diaminobenzidine (Ni-DAB) in the first sequence while TH, PNMT, OT or VP were visualized with DAB alone, resulting in readily distinguishable black and amber reaction products, respectively. 4,4'-Dihydrazino-biphenyl 43-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 8100600-8 1993 Fos was visualized with nickel-intensified diaminobenzidine (Ni-DAB) in the first sequence while TH, PNMT, OT or VP were visualized with DAB alone, resulting in readily distinguishable black and amber reaction products, respectively. ni-dab 61-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 8100600-8 1993 Fos was visualized with nickel-intensified diaminobenzidine (Ni-DAB) in the first sequence while TH, PNMT, OT or VP were visualized with DAB alone, resulting in readily distinguishable black and amber reaction products, respectively. diazobenzenesulfonic acid 64-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 8476611-1 1993 By using spinal cord neurons cultured in chemically defined medium, a double labeling procedure, and blockage with antisense oligonucleotides, we show that induction of c-fos and the subsequent transactivation of the prodynorphin gene are coupled events, triggered by serotonin1A receptor agonists. Oligonucleotides 125-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-174 8487953-3 1993 More complex effects were observed in the neocortex: MK-801 did not influence constitutive expression of different FOS and JUN proteins, but caused marked induction of c-FOS, FOS B, JUN B and JUN D, mainly in layer IV, but also in layers V and VI. Dizocilpine Maleate 53-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-173 19912923-0 1993 Induction and Desensitization of the c-Fos mRNA Response to Nicotine in Rat Brain. Nicotine 60-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 19912923-2 1993 In the present studies, we investigated changes in c-fos mRNA content in several brain regions of the rat in response to nicotine. Nicotine 121-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 19912923-3 1993 A single injection of nicotine ip increased the c-fos mRNA content within 30 min and returned toward baseline by 120 min. Nicotine 22-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 19912923-7 1993 Mecamylamine, a nicotinic cholinergic receptor antagonist, significantly attenuated or eliminated c-fos mRNA response to 1.5 mg/kg nicotine in all regions, except in the cerebellar cortex. Mecamylamine 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 19912923-7 1993 Mecamylamine, a nicotinic cholinergic receptor antagonist, significantly attenuated or eliminated c-fos mRNA response to 1.5 mg/kg nicotine in all regions, except in the cerebellar cortex. Nicotine 131-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 19912923-8 1993 Desensitization of the c-fos mRNA response to nicotine was investigated by administering two injections of 2.0 mg/kg nicotine 2 h apart. Nicotine 46-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 19912923-8 1993 Desensitization of the c-fos mRNA response to nicotine was investigated by administering two injections of 2.0 mg/kg nicotine 2 h apart. Nicotine 117-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 19912923-9 1993 In the hippocampus, dentate gyrus, and piriform cortex, the first dose of nicotine significantly reduced the c-fos mRNA response to a second dose. Nicotine 74-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 19912923-11 1993 In summary, nicotine rapidly elevated the c-fos mRNA content in several rat brain regions. Nicotine 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 8455942-5 1993 In addition, transformation of rat embryo cells induced by an activated ras gene in the presence of the tumor promoter 12-O-tetradecanoyl phorbol 13-acetate (TPA) or by ras plus SV40 large T antigen is also inhibited by this dominant-negative mutant, suggesting that a member of the jun or fos family is involved in the pathways leading to transformation in these systems as well. Tetradecanoylphorbol Acetate 119-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 290-293 8455942-5 1993 In addition, transformation of rat embryo cells induced by an activated ras gene in the presence of the tumor promoter 12-O-tetradecanoyl phorbol 13-acetate (TPA) or by ras plus SV40 large T antigen is also inhibited by this dominant-negative mutant, suggesting that a member of the jun or fos family is involved in the pathways leading to transformation in these systems as well. Tetradecanoylphorbol Acetate 158-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 290-293 8348080-5 1993 Stimulation in the presence of cycloheximide shows that only c-jun, jun-D, c-fos, and JE gene activations are primary responses to the hormone in rat uterine cells. Cycloheximide 31-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 8449605-8 1993 In contrast, fos-specific mRNA and protein in aberrant crypt foci significantly increased when 150 mg/kg all-trans retinoic acid was added to the diet. Tretinoin 115-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 8449605-9 1993 The most important findings of this investigation are that intervention with all-trans retinoic acid in the pre-malignant stage of colon carcinogenesis is effective in decreasing the number and growth of aberrant crypt foci and altering the expression of the c-myc and c-fos genes. Tretinoin 87-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 269-274 8464709-0 1993 Hydrogen peroxide-induced c-fos expression is mediated by arachidonic acid release: role of protein kinase C. We found previously that stimulation of c-fos and c-myc mRNA expression are early events in hydrogen peroxide-induced growth in rat aortic smooth muscle (RASM) cells. Hydrogen Peroxide 0-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 8464709-0 1993 Hydrogen peroxide-induced c-fos expression is mediated by arachidonic acid release: role of protein kinase C. We found previously that stimulation of c-fos and c-myc mRNA expression are early events in hydrogen peroxide-induced growth in rat aortic smooth muscle (RASM) cells. Hydrogen Peroxide 0-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-155 8464709-0 1993 Hydrogen peroxide-induced c-fos expression is mediated by arachidonic acid release: role of protein kinase C. We found previously that stimulation of c-fos and c-myc mRNA expression are early events in hydrogen peroxide-induced growth in rat aortic smooth muscle (RASM) cells. Arachidonic Acid 58-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 8464709-0 1993 Hydrogen peroxide-induced c-fos expression is mediated by arachidonic acid release: role of protein kinase C. We found previously that stimulation of c-fos and c-myc mRNA expression are early events in hydrogen peroxide-induced growth in rat aortic smooth muscle (RASM) cells. Hydrogen Peroxide 202-219 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 8464709-0 1993 Hydrogen peroxide-induced c-fos expression is mediated by arachidonic acid release: role of protein kinase C. We found previously that stimulation of c-fos and c-myc mRNA expression are early events in hydrogen peroxide-induced growth in rat aortic smooth muscle (RASM) cells. Hydrogen Peroxide 202-219 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-155 8464709-1 1993 In the present study, we investigated the role of phospholipase A2 (PLA2) and protein kinase C (PKC) in mediating hydrogen peroxide-induced c-fos mRNA expression in RASM cells. Hydrogen Peroxide 114-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 8464709-2 1993 Mepacrine and p-bromophenacylbromide, potent inhibitors of PLA2 activity, blocked hydrogen peroxide-induced c-fos mRNA expression. Quinacrine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 8464709-2 1993 Mepacrine and p-bromophenacylbromide, potent inhibitors of PLA2 activity, blocked hydrogen peroxide-induced c-fos mRNA expression. 4-bromophenacyl bromide 14-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 8464709-2 1993 Mepacrine and p-bromophenacylbromide, potent inhibitors of PLA2 activity, blocked hydrogen peroxide-induced c-fos mRNA expression. Hydrogen Peroxide 82-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 8464709-3 1993 Arachidonic acid, a product of PLA2 activity, stimulated the expression of c-fos mRNA with a time course similar to that of hydrogen peroxide. Arachidonic Acid 0-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 8464709-4 1993 PKC down-regulation attenuated both hydrogen peroxide and arachidonic acid-induced c-fos mRNA expression by 50%. Arachidonic Acid 58-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 8464709-5 1993 Nordihydroguaiaretic acid (a lipoxygenase-cytochrome P450 monooxygenase inhibitor) significantly inhibited both hydrogen peroxide and arachidonic acid-induced c-fos mRNA expression, whereas indomethacin (a cyclooxygenase inhibitor) had no effect. Masoprocol 0-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-164 8464709-5 1993 Nordihydroguaiaretic acid (a lipoxygenase-cytochrome P450 monooxygenase inhibitor) significantly inhibited both hydrogen peroxide and arachidonic acid-induced c-fos mRNA expression, whereas indomethacin (a cyclooxygenase inhibitor) had no effect. Arachidonic Acid 134-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-164 8464709-6 1993 Together, these findings indicate that 1) hydrogen peroxide-induced c-fos mRNA expression is mediated by PLA2-dependent arachidonic acid release, 2) both PKC-dependent and independent mechanisms are involved in hydrogen peroxide-induced expression of c-fos mRNA and 3) arachidonic acid metabolism via the lipoxygenase-cytochrome P450 monooxygenase pathway appears to be required for hydrogen peroxide-induced expression of c-fos mRNA. Hydrogen Peroxide 42-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 8464709-6 1993 Together, these findings indicate that 1) hydrogen peroxide-induced c-fos mRNA expression is mediated by PLA2-dependent arachidonic acid release, 2) both PKC-dependent and independent mechanisms are involved in hydrogen peroxide-induced expression of c-fos mRNA and 3) arachidonic acid metabolism via the lipoxygenase-cytochrome P450 monooxygenase pathway appears to be required for hydrogen peroxide-induced expression of c-fos mRNA. Hydrogen Peroxide 42-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 251-256 8464709-6 1993 Together, these findings indicate that 1) hydrogen peroxide-induced c-fos mRNA expression is mediated by PLA2-dependent arachidonic acid release, 2) both PKC-dependent and independent mechanisms are involved in hydrogen peroxide-induced expression of c-fos mRNA and 3) arachidonic acid metabolism via the lipoxygenase-cytochrome P450 monooxygenase pathway appears to be required for hydrogen peroxide-induced expression of c-fos mRNA. Hydrogen Peroxide 42-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 251-256 8464709-6 1993 Together, these findings indicate that 1) hydrogen peroxide-induced c-fos mRNA expression is mediated by PLA2-dependent arachidonic acid release, 2) both PKC-dependent and independent mechanisms are involved in hydrogen peroxide-induced expression of c-fos mRNA and 3) arachidonic acid metabolism via the lipoxygenase-cytochrome P450 monooxygenase pathway appears to be required for hydrogen peroxide-induced expression of c-fos mRNA. Arachidonic Acid 120-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 8464709-6 1993 Together, these findings indicate that 1) hydrogen peroxide-induced c-fos mRNA expression is mediated by PLA2-dependent arachidonic acid release, 2) both PKC-dependent and independent mechanisms are involved in hydrogen peroxide-induced expression of c-fos mRNA and 3) arachidonic acid metabolism via the lipoxygenase-cytochrome P450 monooxygenase pathway appears to be required for hydrogen peroxide-induced expression of c-fos mRNA. Arachidonic Acid 120-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 251-256 8464709-6 1993 Together, these findings indicate that 1) hydrogen peroxide-induced c-fos mRNA expression is mediated by PLA2-dependent arachidonic acid release, 2) both PKC-dependent and independent mechanisms are involved in hydrogen peroxide-induced expression of c-fos mRNA and 3) arachidonic acid metabolism via the lipoxygenase-cytochrome P450 monooxygenase pathway appears to be required for hydrogen peroxide-induced expression of c-fos mRNA. Arachidonic Acid 120-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 251-256 8464709-6 1993 Together, these findings indicate that 1) hydrogen peroxide-induced c-fos mRNA expression is mediated by PLA2-dependent arachidonic acid release, 2) both PKC-dependent and independent mechanisms are involved in hydrogen peroxide-induced expression of c-fos mRNA and 3) arachidonic acid metabolism via the lipoxygenase-cytochrome P450 monooxygenase pathway appears to be required for hydrogen peroxide-induced expression of c-fos mRNA. Hydrogen Peroxide 211-228 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 8464709-6 1993 Together, these findings indicate that 1) hydrogen peroxide-induced c-fos mRNA expression is mediated by PLA2-dependent arachidonic acid release, 2) both PKC-dependent and independent mechanisms are involved in hydrogen peroxide-induced expression of c-fos mRNA and 3) arachidonic acid metabolism via the lipoxygenase-cytochrome P450 monooxygenase pathway appears to be required for hydrogen peroxide-induced expression of c-fos mRNA. Hydrogen Peroxide 211-228 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 251-256 8464709-6 1993 Together, these findings indicate that 1) hydrogen peroxide-induced c-fos mRNA expression is mediated by PLA2-dependent arachidonic acid release, 2) both PKC-dependent and independent mechanisms are involved in hydrogen peroxide-induced expression of c-fos mRNA and 3) arachidonic acid metabolism via the lipoxygenase-cytochrome P450 monooxygenase pathway appears to be required for hydrogen peroxide-induced expression of c-fos mRNA. Hydrogen Peroxide 211-228 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 251-256 8464709-6 1993 Together, these findings indicate that 1) hydrogen peroxide-induced c-fos mRNA expression is mediated by PLA2-dependent arachidonic acid release, 2) both PKC-dependent and independent mechanisms are involved in hydrogen peroxide-induced expression of c-fos mRNA and 3) arachidonic acid metabolism via the lipoxygenase-cytochrome P450 monooxygenase pathway appears to be required for hydrogen peroxide-induced expression of c-fos mRNA. Arachidonic Acid 269-285 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 8464709-6 1993 Together, these findings indicate that 1) hydrogen peroxide-induced c-fos mRNA expression is mediated by PLA2-dependent arachidonic acid release, 2) both PKC-dependent and independent mechanisms are involved in hydrogen peroxide-induced expression of c-fos mRNA and 3) arachidonic acid metabolism via the lipoxygenase-cytochrome P450 monooxygenase pathway appears to be required for hydrogen peroxide-induced expression of c-fos mRNA. Arachidonic Acid 269-285 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 251-256 8464709-6 1993 Together, these findings indicate that 1) hydrogen peroxide-induced c-fos mRNA expression is mediated by PLA2-dependent arachidonic acid release, 2) both PKC-dependent and independent mechanisms are involved in hydrogen peroxide-induced expression of c-fos mRNA and 3) arachidonic acid metabolism via the lipoxygenase-cytochrome P450 monooxygenase pathway appears to be required for hydrogen peroxide-induced expression of c-fos mRNA. Arachidonic Acid 269-285 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 251-256 8464709-6 1993 Together, these findings indicate that 1) hydrogen peroxide-induced c-fos mRNA expression is mediated by PLA2-dependent arachidonic acid release, 2) both PKC-dependent and independent mechanisms are involved in hydrogen peroxide-induced expression of c-fos mRNA and 3) arachidonic acid metabolism via the lipoxygenase-cytochrome P450 monooxygenase pathway appears to be required for hydrogen peroxide-induced expression of c-fos mRNA. Hydrogen Peroxide 211-228 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 8464709-6 1993 Together, these findings indicate that 1) hydrogen peroxide-induced c-fos mRNA expression is mediated by PLA2-dependent arachidonic acid release, 2) both PKC-dependent and independent mechanisms are involved in hydrogen peroxide-induced expression of c-fos mRNA and 3) arachidonic acid metabolism via the lipoxygenase-cytochrome P450 monooxygenase pathway appears to be required for hydrogen peroxide-induced expression of c-fos mRNA. Hydrogen Peroxide 211-228 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 251-256 8464709-6 1993 Together, these findings indicate that 1) hydrogen peroxide-induced c-fos mRNA expression is mediated by PLA2-dependent arachidonic acid release, 2) both PKC-dependent and independent mechanisms are involved in hydrogen peroxide-induced expression of c-fos mRNA and 3) arachidonic acid metabolism via the lipoxygenase-cytochrome P450 monooxygenase pathway appears to be required for hydrogen peroxide-induced expression of c-fos mRNA. Hydrogen Peroxide 211-228 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 251-256 8469433-1 1993 The effects of high-dose ketamine on the c-fos protein (c-Fos) expression were investigated in rat by an immunohistochemical technique. Ketamine 25-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 8469433-1 1993 The effects of high-dose ketamine on the c-fos protein (c-Fos) expression were investigated in rat by an immunohistochemical technique. Ketamine 25-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 8510493-2 1993 The effect of the two chemical convulsants, pentylenetetrazol (PTZ) and flurothyl, on the spatial and temporal pattern of c-fos-like immunoreactivity in immature (postnatal day (P) 10) was compared to that in adult rats. Pentylenetetrazole 44-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 8510493-2 1993 The effect of the two chemical convulsants, pentylenetetrazol (PTZ) and flurothyl, on the spatial and temporal pattern of c-fos-like immunoreactivity in immature (postnatal day (P) 10) was compared to that in adult rats. Pentylenetetrazole 63-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 8510493-4 1993 C-fos-like immunoreactivity was examined at 2, 4, and 6 h after onset of chemically induced seizures or O2 deprivation at both ages. Oxygen 104-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8510500-4 1993 The induction of c-Jun, Jun-B, Jun-D and Fos-related proteins was prevented by administration of an N-methyl-D-aspartate receptor antagonist, which also blocked LTP induction, and by pentobarbital, which reduced but did not block LTP. Pentobarbital 183-196 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 8492912-6 1993 Pretreatment of animals with the serotonin2/1C receptor antagonist ritanserin (0.4 mg/kg) markedly attenuated Fos expression in all reactive areas of the brain. Ritanserin 67-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 8492912-8 1993 Spiperone (1 mg/kg), a mixed serotonin2 and dopamine D2 antagonist, also attenuated the Fos response in the same regions, but had the effect of inducing Fos expression on its own in other extrapyramidal brain regions. Spiperone 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 8492912-8 1993 Spiperone (1 mg/kg), a mixed serotonin2 and dopamine D2 antagonist, also attenuated the Fos response in the same regions, but had the effect of inducing Fos expression on its own in other extrapyramidal brain regions. Spiperone 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-156 8098512-4 1993 We have examined the expression of immediate early genes in central neurons of the rat and now show that a 6-hydroxydopamine-induced axotomy of the dopaminergic nigrostriatal pathway results in a substantial increase in the levels of c-jun (but not c-fos) messenger RNA and protein within neurons of the substantia nigra pars compacta. Oxidopamine 107-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 249-254 8461977-0 1993 Effect of locus coeruleus lesion on c-fos expression in the cerebral cortex caused by yohimbine injection or stress. Yohimbine 86-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 8387656-6 1993 c-fos immunoreactivity studies showed that ingestion of saccharin induced a remarkable activation of the central lateral (cl) subnucleus of the PBN in normal rats, however, rats with the CTA to saccharin showed c-fos neurons in the ventral lateral (vl) subnucleus of the PBN. Saccharin 56-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8387656-6 1993 c-fos immunoreactivity studies showed that ingestion of saccharin induced a remarkable activation of the central lateral (cl) subnucleus of the PBN in normal rats, however, rats with the CTA to saccharin showed c-fos neurons in the ventral lateral (vl) subnucleus of the PBN. Saccharin 56-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 211-216 8461977-1 1993 The injection of the alpha-2 adrenoceptor antagonist, yohimbine, has been shown to increase c-fos immunoreactivity in the rat cerebral cortex. Yohimbine 54-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 8461977-5 1993 It is concluded that the noradrenergic system plays a necessary role in the above c-fos responses in the cortex to yohimbine and to stress. Yohimbine 115-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 8384296-4 1993 This accumulation of c-fos mRNA by myocyte stretching was inhibited markedly by down-regulation of protein kinase C. Moreover, myocyte stretching increased inositol phosphate levels. Inositol Phosphates 156-174 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 8457843-5 1993 Blockade of the effects of progesterone or removal of the pups for 24 h, resulted in a moderate level of cFos intensity in response to NMA. nma 135-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-109 8381969-8 1993 Mutational analysis of the steroid hormone binding domain and the overlapping AP-1 site at the VDRE supports mutually exclusive occupancy by Fos-Jun heterodimers and vitamin D receptor. Steroids 27-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 8381969-9 1993 Such protein-DNA interactions at the VDRE are consistent with repression of competency for vitamin D-mediated transcriptional enhancement in proliferating osteoblasts expressing high levels of Fos and Jun. Vitamin D 91-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-196 8453468-0 1993 Sensitization of c-fos expression in rat striatum following multiple challenges with D-amphetamine. Dextroamphetamine 85-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 8453468-1 1993 D-Amphetamine transiently stimulates the expression of the immediate-early response gene, c-fos, in rat striatal cell nuclei. Dextroamphetamine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 8453468-3 1993 induced a significantly greater expression of Fos-like immunoreactivity in striatum of rats treated three days previously with D-amphetamine compared to rats treated three days previously with saline. Dextroamphetamine 127-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 8453468-5 1993 This sensitization of c-fos expression following a repeated administration of D-amphetamine indicates an increased activation of post-synaptic elements in rat striatum. Dextroamphetamine 78-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 8453476-2 1993 A massive and transitory increase in c-fos mRNA and Fos protein occurred in rats intoxicated by a single dose of soman (organophosphate compound and irreversible cholinesterase inhibitor) only in animals that had seizures. Organophosphates 120-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 8095432-0 1993 Induction of c-fos immunoreactivity in tyrosine hydroxylase and phenylethanolamine-N-methyltransferase immunoreactive neurons of the medulla oblongata of the rat after phosphate-buffered saline load in the urethane-anaesthetized rat. Phosphate-Buffered Saline 168-193 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 8095432-0 1993 Induction of c-fos immunoreactivity in tyrosine hydroxylase and phenylethanolamine-N-methyltransferase immunoreactive neurons of the medulla oblongata of the rat after phosphate-buffered saline load in the urethane-anaesthetized rat. Urethane 206-214 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 8095432-1 1993 We have studied the induction of c-fos immunoreactivity (c-fos IR) in catecholaminergic and vasopressinergic immunoreactive neurons after repeated phosphate-buffered saline (PBS) loading or after repeated elicitation of the baroreceptor reflex via repeated infusion of the vasoconstrictor agent L-phenylephrine. Phosphate-Buffered Saline 147-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 8095432-1 1993 We have studied the induction of c-fos immunoreactivity (c-fos IR) in catecholaminergic and vasopressinergic immunoreactive neurons after repeated phosphate-buffered saline (PBS) loading or after repeated elicitation of the baroreceptor reflex via repeated infusion of the vasoconstrictor agent L-phenylephrine. pbs 174-177 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 8095432-1 1993 We have studied the induction of c-fos immunoreactivity (c-fos IR) in catecholaminergic and vasopressinergic immunoreactive neurons after repeated phosphate-buffered saline (PBS) loading or after repeated elicitation of the baroreceptor reflex via repeated infusion of the vasoconstrictor agent L-phenylephrine. pbs 174-177 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 8469394-1 1993 Recent studies have shown that the centrally active muscarinic agonist pilocarpine induces c-fos mRNA in rat cortex. Pilocarpine 71-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 8469394-2 1993 Here we describe the localization of muscarinic receptor-induced FOS protein, within the rat central nervous system (CNS), following administration of pilocarpine (25 mg/kg). Pilocarpine 151-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 8469394-6 1993 Both atropine (10 mg/kg) and pirenzepine (100 mg/kg) reduced FOS induction suggesting that a pirenzepine-sensitive muscarinic receptor was involved. Atropine 5-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 8469394-6 1993 Both atropine (10 mg/kg) and pirenzepine (100 mg/kg) reduced FOS induction suggesting that a pirenzepine-sensitive muscarinic receptor was involved. Pirenzepine 29-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 8381576-0 1993 Manidipine inhibits endothelin-1-induced [Ca2+]i signaling but potentiates endothelin"s effect on c-fos and c-jun induction in vascular smooth muscle and glomerular mesangial cells. manidipine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 8381576-6 1993 In contrast, manidipine (10(-5) mol/L) potentiated ET-1-induced c-fos and c-jun expression in A7r5 cells. manidipine 13-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 8385579-9 1993 The investigations have focused on the Fos-Jun family of immediate early gene transcription factors, and the CREB family of transcription factors, as possible mediators of the effects of chronic opiate and cocaine exposure on regulation of neuronal gene expression. Opiate Alkaloids 195-201 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 8426221-0 1993 Coordinate expression of c-fos and jun B is induced in the rat striatum by cocaine. Cocaine 75-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 8426221-2 1993 In the present study, we tested for such selectivity of stimulated gene expression by monitoring the expression of fos/jun gene mRNAs in the striatum in rats treated in vivo with the indirect dopamine agonist cocaine. Cocaine 209-216 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-118 8426221-3 1993 We found by Northern blot and in situ hybridization analysis that cocaine induces the coordinate expression of c-fos and jun B mRNAs in neurons of the rat"s striatum. Cocaine 66-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 8426221-5 1993 With the same probe, we could detect the induction of c-jun mRNA (as well as that of c-fos and jun B mRNAs) in the hippocampus following administration of pentylenetetrazol. Pentylenetetrazole 155-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 8426221-6 1993 The induction of expression of c-fos and jun B was rapid and transient, with peak expression occurring at approximately 1 hr after cocaine administration, and the induction of the two genes was in similar striatal sites. Cocaine 131-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 8426221-7 1993 These results establish that differential patterns of expression of fos/jun genes occur in striatal neurons following exposure to cocaine, a potent psychomotor stimulant. Cocaine 130-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 8448663-1 1993 Application of potassium chloride (KCl) to the brain surface elicits spreading depression which leads to a marked induction of the proto-oncogene c-fos in the treated cerebral cortex at the earliest time examined (90 min). Potassium Chloride 15-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-151 8448663-1 1993 Application of potassium chloride (KCl) to the brain surface elicits spreading depression which leads to a marked induction of the proto-oncogene c-fos in the treated cerebral cortex at the earliest time examined (90 min). Potassium Chloride 35-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-151 8448663-2 1993 High levels of c-fos immunoreactivity are observed up to 6 h after KCl treatment. Potassium Chloride 67-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 8421668-5 1993 The c-Fos peptide was phosphorylated at a serine corresponding to Ser362, a site critically implicated in the capacity of c-Fos to exhibit transrepressive activity [Ofir, R., Dwarki, V. J., Rashid, D. & Verma, I. M. (1990) Nature (London) 348, 80-82)]. Serine 42-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 8421668-5 1993 The c-Fos peptide was phosphorylated at a serine corresponding to Ser362, a site critically implicated in the capacity of c-Fos to exhibit transrepressive activity [Ofir, R., Dwarki, V. J., Rashid, D. & Verma, I. M. (1990) Nature (London) 348, 80-82)]. Serine 42-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 7682304-0 1993 The induction and suppression of c-fos expression in the rat brain by cholecystokinin and its antagonist L364,718. 3-(4-chlorophenyl)pentanedioic acid 105-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 7682304-4 1993 The c-fos expression in these areas was suppressed by the administration of L364,718 (120 micrograms/kg). 3-(4-chlorophenyl)pentanedioic acid 76-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 7748323-1 1993 The expression of c-fos mRNA in rat brain was induced by intraperitoneal (ip) administration of N-methyl-D-aspartate (NMDA) and kainic acid, agonists of different classes of glutamate receptors and by caffeine, an antagonist of adenosine receptors. N-Methylaspartate 96-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 7748323-1 1993 The expression of c-fos mRNA in rat brain was induced by intraperitoneal (ip) administration of N-methyl-D-aspartate (NMDA) and kainic acid, agonists of different classes of glutamate receptors and by caffeine, an antagonist of adenosine receptors. N-Methylaspartate 118-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 7748323-1 1993 The expression of c-fos mRNA in rat brain was induced by intraperitoneal (ip) administration of N-methyl-D-aspartate (NMDA) and kainic acid, agonists of different classes of glutamate receptors and by caffeine, an antagonist of adenosine receptors. Kainic Acid 128-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 7748323-1 1993 The expression of c-fos mRNA in rat brain was induced by intraperitoneal (ip) administration of N-methyl-D-aspartate (NMDA) and kainic acid, agonists of different classes of glutamate receptors and by caffeine, an antagonist of adenosine receptors. Caffeine 201-209 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 7748323-4 1993 However, ethanol-withdrawn rats showed a marked increase in c-fos expression. Ethanol 9-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 7748323-7 1993 The withdrawal-induced expression of c-fos mRNA could be inhibited by prior administration of MK801. Dizocilpine Maleate 94-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 8420627-0 1993 Cocaine-induced expression of striatal c-fos in the rat is inhibited by NMDA receptor antagonists. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 8420627-4 1993 Pretreatment with MK-801 (1 mg/kg) or CPP (5 mg/kg) also interfered, albeit to a lesser extent, with the expression of c-fos by cocaine in awake animals. Dizocilpine Maleate 18-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 8420627-4 1993 Pretreatment with MK-801 (1 mg/kg) or CPP (5 mg/kg) also interfered, albeit to a lesser extent, with the expression of c-fos by cocaine in awake animals. Cocaine 128-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 8420627-5 1993 These results indicate that cocaine induction of cellular c-fos in the caudate putamen is mediated at least in part by NMDA-sensitive receptors. Cocaine 28-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 8453494-0 1993 Dexfenfluramine induces Fos-like immunoreactivity in discrete brain regions in rats. Dexfenfluramine 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 8453494-1 1993 Administration of the anorectic agent, dexfenfluramine (DFEN) to rats induced dose- and time-dependent expression of Fos-like immunoreactivity in several discrete brain regions of rats. Dexfenfluramine 39-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 8453494-1 1993 Administration of the anorectic agent, dexfenfluramine (DFEN) to rats induced dose- and time-dependent expression of Fos-like immunoreactivity in several discrete brain regions of rats. Dexfenfluramine 56-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 8453494-4 1993 Rats that were made hungry by either food deprivation or administration of insulin also showed induction of Fos in several brain regions, but only that in the supramamillary/ventral tegmental area was suppressed by pretreatment with an anorectic dose of DFEN. Dexfenfluramine 254-258 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 7904879-3 1993 The 34-40 kDa proteins (CREB-327/341) were able to bind to the CRE of cAMP-inducible genes (somatostatin, c-fos, E2A), but not to genes whose expression is not controlled by cAMP (glucagon, parathyroid hormone). Cyclic AMP 70-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 7904879-3 1993 The 34-40 kDa proteins (CREB-327/341) were able to bind to the CRE of cAMP-inducible genes (somatostatin, c-fos, E2A), but not to genes whose expression is not controlled by cAMP (glucagon, parathyroid hormone). Cyclic AMP 174-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 8385579-9 1993 The investigations have focused on the Fos-Jun family of immediate early gene transcription factors, and the CREB family of transcription factors, as possible mediators of the effects of chronic opiate and cocaine exposure on regulation of neuronal gene expression. Cocaine 206-213 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 8419141-7 1993 Estradiol was shown to stimulate IEC-6 cell c-fos mRNA content rapidly and transiently in a manner analogous to that which has been previously demonstrated for other estrogen-responsive tissues. Estradiol 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 8469783-1 1993 This study was designed to investigate the influence of intracellular ionized calcium ([Ca2+]i) on the induction of c-fos, c-jun, c-myc, and hsp70 genes after oxidant stress induced by xanthine/xanthine oxidase (X/XOD) treatment or after heat shock using primary cultures of rat proximal tubule epithelium (PTE). Calcium 78-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 8325038-6 1993 Recent observations implying reactive oxygen as the transduction signal that mediates activation of c-fos and c-jun genes are presently considered to provide an explanation for the induction of GST gene expression by chemical agents of diverse structure. Oxygen 38-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 8395405-8 1993 C-Fos expression as determined by immunoreactivity for the nuclear phosphoprotein (Fos) indicated neuronal activation at NMDA infusion sites, in the perirhinal cortex, hippocampus, and other sites throughout the injected hemisphere. N-Methylaspartate 121-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 8395405-8 1993 C-Fos expression as determined by immunoreactivity for the nuclear phosphoprotein (Fos) indicated neuronal activation at NMDA infusion sites, in the perirhinal cortex, hippocampus, and other sites throughout the injected hemisphere. N-Methylaspartate 121-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 2-5 8417145-4 1993 The activation of protein kinase C appears as an obligatory step during these processes, because (a) inhibition of protein kinase C by staurosporine blocks the induction by endothelin or phorbol esters of both c-fos and nerve growth factor, and (b) phorbol ester-evoked down-regulation of protein kinase C completely abolishes the c-fos induction by endothelin, but not that by the beta-adrenergic agonist isoproterenol, a known activator of the cyclic AMP-dependent pathway. Staurosporine 135-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 210-215 8417145-4 1993 The activation of protein kinase C appears as an obligatory step during these processes, because (a) inhibition of protein kinase C by staurosporine blocks the induction by endothelin or phorbol esters of both c-fos and nerve growth factor, and (b) phorbol ester-evoked down-regulation of protein kinase C completely abolishes the c-fos induction by endothelin, but not that by the beta-adrenergic agonist isoproterenol, a known activator of the cyclic AMP-dependent pathway. Staurosporine 135-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 331-336 8417145-4 1993 The activation of protein kinase C appears as an obligatory step during these processes, because (a) inhibition of protein kinase C by staurosporine blocks the induction by endothelin or phorbol esters of both c-fos and nerve growth factor, and (b) phorbol ester-evoked down-regulation of protein kinase C completely abolishes the c-fos induction by endothelin, but not that by the beta-adrenergic agonist isoproterenol, a known activator of the cyclic AMP-dependent pathway. Phorbol Esters 187-201 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 210-215 8417145-4 1993 The activation of protein kinase C appears as an obligatory step during these processes, because (a) inhibition of protein kinase C by staurosporine blocks the induction by endothelin or phorbol esters of both c-fos and nerve growth factor, and (b) phorbol ester-evoked down-regulation of protein kinase C completely abolishes the c-fos induction by endothelin, but not that by the beta-adrenergic agonist isoproterenol, a known activator of the cyclic AMP-dependent pathway. Phorbol Esters 187-200 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 210-215 8453036-1 1993 Using a selective monoclonal Fos antibody, we have studied the expression of the c-fos proto-oncogene in the olfactory bulb of awake rats stimulated with propionic acid vapours. propionic acid 154-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 8453036-4 1993 We observed that the low level of basal c-fos expression raised significantly under olfactory stimulation in the bulbar column defined by the foci of high 2-deoxyglucose glomerular uptake. Deoxyglucose 155-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 8446434-0 1993 Effects of morphine and naloxone on basal and evoked Fos-like immunoreactivity in lumbar spinal cord neurons of arthritic rats. Naloxone 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 8446434-3 1993 The aim of this study was to evaluate Fos-LI following morphine injection (1) in arthritic animals in the absence of any stimulation, (2) in arthritic rats after ankle stimulation pretreated with morphine or by the combination of morphine and naloxone, and (3) following naloxone treatment in non-stimulated and stimulated polyarthritic rats. Morphine 55-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 8446434-3 1993 The aim of this study was to evaluate Fos-LI following morphine injection (1) in arthritic animals in the absence of any stimulation, (2) in arthritic rats after ankle stimulation pretreated with morphine or by the combination of morphine and naloxone, and (3) following naloxone treatment in non-stimulated and stimulated polyarthritic rats. Naloxone 243-251 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 8446434-3 1993 The aim of this study was to evaluate Fos-LI following morphine injection (1) in arthritic animals in the absence of any stimulation, (2) in arthritic rats after ankle stimulation pretreated with morphine or by the combination of morphine and naloxone, and (3) following naloxone treatment in non-stimulated and stimulated polyarthritic rats. Naloxone 271-279 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 8446434-7 1993 In contrast, Fos-LI evoked by noxious pressure was strongly depressed by morphine. Morphine 73-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 8446434-8 1993 In the superficial laminae pretreatment with a single morphine injection of either 0.5 or 1 mg/kg, i.v., reduced by more than 50% the number of Fos-LI neurons and at 3 mg/kg completely abolished the labeling evoked by the stimulation. Morphine 54-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-147 1291036-0 1992 Histamine-caused itch induces Fos-like immunoreactivity in dorsal horn neurons: effect of morphine pretreatment. Histamine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 1291036-4 1992 After 2 h, the number of neurons showing Fos-like immunoreactivity was significantly increased in a dose-dependent manner in the ipsilateral dorsal horn when compared to saline-treated and scratched controls. Sodium Chloride 170-176 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 1291036-6 1992 Expression of Fos-like immunoreactivity in these animals was markedly reduced by morphine pretreatment (10 mg/kg, i.p. Morphine 81-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 1335693-2 1992 Stimulation of beta-adrenergic receptors by isoproterenol, or addition of 8-bromoadenosine 3",5"-cyclic monophosphate, induces the expression of c-fos and jun B by a protein kinase A-mediated pathway. Isoproterenol 44-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-150 1335693-2 1992 Stimulation of beta-adrenergic receptors by isoproterenol, or addition of 8-bromoadenosine 3",5"-cyclic monophosphate, induces the expression of c-fos and jun B by a protein kinase A-mediated pathway. 8-Bromo Cyclic Adenosine Monophosphate 74-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-150 1335693-5 1992 Increases in cytosolic Ca2+ by A23187 or ionomycin induce only the expression of c-fos gene. Calcimycin 31-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 1335693-5 1992 Increases in cytosolic Ca2+ by A23187 or ionomycin induce only the expression of c-fos gene. Ionomycin 41-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 1332847-3 1992 Other agents that increase c-fos mRNA levels and DNA synthesis in FRTL-5 cells include TSH, insulin, insulin-like growth factor-I, phorbol esters, A23187, and alpha 1-adrenergic agents; the last two agents also act by increasing cytosolic Ca2+ levels. Thyrotropin 87-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 1332847-3 1992 Other agents that increase c-fos mRNA levels and DNA synthesis in FRTL-5 cells include TSH, insulin, insulin-like growth factor-I, phorbol esters, A23187, and alpha 1-adrenergic agents; the last two agents also act by increasing cytosolic Ca2+ levels. Calcimycin 147-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 1464772-3 1992 However, chemical irritation with 1% acetic acid or mechanical stimulation of the LUT markedly increased the number of c-fos-positive neurons (56-180 cells/L6 section) in four regions of the caudal lumbosacral (L6-S1) spinal cord: medial dorsal horn (MDH), lateral dorsal horn, dorsal commissure (DCM), and sacral parasympathetic nucleus (SPN). Acetic Acid 37-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 1361576-7 1992 These findings demonstrate that regionally enhanced electrophysiological sensitivity of striatal neurons to D1-DA receptor agonists after neonatal 6-OHDA-induced lesions is associated with regional changes in c-fos-like immunoreactivity and tyrosine hydroxylase-like immunohistochemistry, but not with changes in D1-DA receptor autoradiography or D1-DA-stimulated adenylyl cyclase activity. Oxidopamine 147-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 209-214 1337282-0 1992 C-fos expression in the rat brain after intraperitoneal injection of lithium chloride. Lithium Chloride 69-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 1337282-1 1992 The distribution of evoked expression of the proto-oncogene c-fos was immunohistochemically examined in the rat brain after intraperitoneal injection of isotonic LiCl, which is commonly used to induce internal malaise in the conditioned taste aversion paradigm. Lithium Chloride 162-166 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-65 1338729-10 1992 Analogous effects were observed with TPA which minimally stimulated c-fos and c-jun mRNAs on day 0, but markedly increased these messages on day 2. Tetradecanoylphorbol Acetate 37-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 1338729-11 1992 These studies demonstrate that c-fos and c-jun mRNAs can be induced in cultured rat granulosa cells by acute gonadotropin, (Bu)2cAMP or phorbol ester treatment and suggest that these immediate early proto-oncogenes may play a role in granulosa cell function. Phorbol Esters 136-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 1488127-0 1992 Regional expression of c-fos antigen in the basal forebrain following intraventricular infusions of angiotensin and its modulation by drinking either water or saline. Water 150-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 1488127-0 1992 Regional expression of c-fos antigen in the basal forebrain following intraventricular infusions of angiotensin and its modulation by drinking either water or saline. Sodium Chloride 159-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 1488127-9 1992 When water was withheld for 15 min, but then allowed, rats drank the same total volume but c-fos expression was no longer inhibited in either the supraoptic nucleus or paraventricular nucleus. Water 5-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 1488127-12 1992 There was a marked correlation between the number of c-fos-positive neurons in the supraoptic nucleus or paraventricular nucleus and plasma levels of corticosterone 60 min after infusion, but not with arginine-vasopressin levels. Corticosterone 150-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 1445353-2 1992 In the present study, we investigated the effects of genistein, a tyrosine kinase inhibitor, and NiCl2, a Ca2+ influx blocker, on PDGF-induced Ca2+ transient and on expression of c-fos and c-myc mRNA. nickel chloride 97-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-184 1445353-7 1992 In the presence of 10 microM genistein and 1 mM NiCl2, PDGF induced c-fos and c-myc mRNA, although the [Ca2+]i elevation could be completely blocked by these two agents. Genistein 29-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 1445353-7 1992 In the presence of 10 microM genistein and 1 mM NiCl2, PDGF induced c-fos and c-myc mRNA, although the [Ca2+]i elevation could be completely blocked by these two agents. nickel chloride 48-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 1281256-0 1992 Administration of quinolinic acid in the rat hippocampus induces expression of c-fos and NGFI-A. Quinolinic Acid 18-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 1281256-1 1992 We have studied the effect of intrahippocampal administration of quinolinic acid (QUIN) on the temporal expression of mRNAs encoding the immediate early genes (IEGs) c-fos and NGFI-A, by in situ hybridization histochemistry. Quinolinic Acid 65-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-171 1281256-1 1992 We have studied the effect of intrahippocampal administration of quinolinic acid (QUIN) on the temporal expression of mRNAs encoding the immediate early genes (IEGs) c-fos and NGFI-A, by in situ hybridization histochemistry. Quinolinic Acid 82-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-171 1281256-10 1992 Twelve h after QUIN administration, c-fos and NGFI-A mRNAs were present in the dentate gyrus. Quinolinic Acid 15-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 1334199-0 1992 Melatonin influences Fos expression in the rat suprachiasmatic. Melatonin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 1334199-1 1992 Administration of the pineal hormone melatonin to rats induces expression of Fos, the protein product of the c-fos proto-oncogene, in the suprachiasmatic nucleus (SCN), the putative biological clock of mammals. Melatonin 37-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-80 1334199-1 1992 Administration of the pineal hormone melatonin to rats induces expression of Fos, the protein product of the c-fos proto-oncogene, in the suprachiasmatic nucleus (SCN), the putative biological clock of mammals. Melatonin 37-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 1422856-0 1992 Induction of c-fos immunostaining in the rat brain after the systemic administration of nicotine. Nicotine 88-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 1422856-1 1992 To search for evidence of altered neuronal gene expression in response to exposure to the highly addictive drug nicotine, rat brains were examined by immunocytochemistry for the fos protein after the systemic administration of nicotine. Nicotine 112-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 178-181 1422856-2 1992 The drug was administered as an IV infusion over 1 h At a dose of 2 mg/kg, the most dramatic nicotine-induced fos nuclear immunostaining was seen in central visual pathways, including the superficial superior colliculus and the medial terminal nu. Nicotine 93-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 1422856-5 1992 A minimal increase in fos immunostaining was seen after a nicotine dose of 0.5 mg/kg, with a much greater response after 1 or 2 mg/kg. Nicotine 58-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-25 1422856-7 1992 c-fos expression was substantially attenuated in the superficial gray layer of superior colliculus, medial terminal nucleus of the accessory optic tract, and the interpeduncular nucleus by pretreatment with the centrally acting nicotine antagonist mecamylamine, 5 mg/kg IP, but not with the peripherally acting antagonist hexamethonium, 4 mg/kg IP. Nicotine 228-236 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 1422856-7 1992 c-fos expression was substantially attenuated in the superficial gray layer of superior colliculus, medial terminal nucleus of the accessory optic tract, and the interpeduncular nucleus by pretreatment with the centrally acting nicotine antagonist mecamylamine, 5 mg/kg IP, but not with the peripherally acting antagonist hexamethonium, 4 mg/kg IP. Mecamylamine 248-260 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 1422856-7 1992 c-fos expression was substantially attenuated in the superficial gray layer of superior colliculus, medial terminal nucleus of the accessory optic tract, and the interpeduncular nucleus by pretreatment with the centrally acting nicotine antagonist mecamylamine, 5 mg/kg IP, but not with the peripherally acting antagonist hexamethonium, 4 mg/kg IP. Hexamethonium 322-335 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 1422856-8 1992 These observations identify a subset of central nervous system neurons that respond to nicotine with altered expression of the immediate early gene c-fos. Nicotine 87-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 1330491-7 1992 Northern blot analysis of mRNA from UMR 106-01 cells treated for 3 h with 2 microM PGE2, 10 nM PTH, or 10 ng/ml EGF in the presence of cycloheximide demonstrated that all three agents induced the expression of c-fos and c-jun mRNA. Cycloheximide 135-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 210-215 1330491-10 1992 c-fos mRNA was induced by treatment with 50 ng/ml tetradecanoyl phorbol acetate or by 40 ng/ml forskolin, while induction of Egr-1 mRNA was stimulated by treatment with tetradecanoyl phorbol acetate, but not forskolin. Tetradecanoylphorbol Acetate 50-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 1330491-10 1992 c-fos mRNA was induced by treatment with 50 ng/ml tetradecanoyl phorbol acetate or by 40 ng/ml forskolin, while induction of Egr-1 mRNA was stimulated by treatment with tetradecanoyl phorbol acetate, but not forskolin. Colforsin 95-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 1425409-2 1992 In female rats, a steroid-induced LH surge is accompanied by an increase in FOS-positive GnRH neurons, especially in the region of the organum vasculosum of the lamina terminalis. Steroids 18-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 1385204-10 1992 Both systemic and intracoerulear soman administration completely inhibited AChE staining in LC and rapidly induced the expression of c-fos in LC neurons. Soman 33-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 8469783-3 1993 The expression of both c-fos following X/XOD treatment and hsp70 following heat shock was markedly decreased through chelation of [Ca2+]i by Quin 2/AM. Quin2 141-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 8469783-4 1993 The c-fos expression following X/XOD treatment was partly reduced by a protein kinase C inhibitor, staurosporine (ST), and markedly inhibited by another protein kinase inhibitor, 2-aminopurine (2AP), while both ST and 2AP markedly reduced hsp70 expression. Staurosporine 99-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 8469783-4 1993 The c-fos expression following X/XOD treatment was partly reduced by a protein kinase C inhibitor, staurosporine (ST), and markedly inhibited by another protein kinase inhibitor, 2-aminopurine (2AP), while both ST and 2AP markedly reduced hsp70 expression. Staurosporine 114-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 8469783-4 1993 The c-fos expression following X/XOD treatment was partly reduced by a protein kinase C inhibitor, staurosporine (ST), and markedly inhibited by another protein kinase inhibitor, 2-aminopurine (2AP), while both ST and 2AP markedly reduced hsp70 expression. 2-Aminopurine 179-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 1465198-13 1992 Corticosterone levels were raised by both stress and corticotropin-releasing factor, but pretreatment with alpha-helical corticotropin-releasing factor reduced them after either procedure, which correlates with c-fos expression in the paraventricular nucleus and ventrolateral septum. Corticosterone 0-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 211-216 1480173-9 1992 8-Bromo-cAMP and phorbol 12-myristate 13-acetate (PMA) caused increases in the abundance of c-fos and c-jun transcripts. 8-Bromo Cyclic Adenosine Monophosphate 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 1480173-9 1992 8-Bromo-cAMP and phorbol 12-myristate 13-acetate (PMA) caused increases in the abundance of c-fos and c-jun transcripts. Tetradecanoylphorbol Acetate 17-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 1480173-9 1992 8-Bromo-cAMP and phorbol 12-myristate 13-acetate (PMA) caused increases in the abundance of c-fos and c-jun transcripts. Tetradecanoylphorbol Acetate 50-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 1450955-4 1992 Fos immunoreactivity was induced in cells in several medullary, hypothalamic and limbic structures with infusions of angiotensin II or phenylephrine at pressor doses. Phenylephrine 135-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 1467932-0 1992 The 5-hydroxytryptamine agonist fenfluramine increases Fos-like immunoreactivity in the brain. Fenfluramine 32-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 1467932-1 1992 This study was designed to assess the effects of the 5-hydroxytryptamine (5-HT) indirect agonist fenfluramine on the brain distribution of Fos- and corticotropin-releasing factor-like immunoreactivity (F-LI and CRF-LI). Serotonin 53-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 1467932-1 1992 This study was designed to assess the effects of the 5-hydroxytryptamine (5-HT) indirect agonist fenfluramine on the brain distribution of Fos- and corticotropin-releasing factor-like immunoreactivity (F-LI and CRF-LI). Bufotenin 76-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 1467932-1 1992 This study was designed to assess the effects of the 5-hydroxytryptamine (5-HT) indirect agonist fenfluramine on the brain distribution of Fos- and corticotropin-releasing factor-like immunoreactivity (F-LI and CRF-LI). Fenfluramine 97-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 1467932-10 1992 This study provides evidence for an involvement of the immediate-early genes c-fos in the central action of fenfluramine. Fenfluramine 108-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 1458316-4 1992 Intravenous infusion of the vasodilating agent, nitroprusside, which lowered the blood pressure to levels comparable to that attained by hemorrhage, induced Fos-IR in greater than 65% of AVP-containing neurons in the SON, while relatively few AVP neurons in the PVN were Fos positive. Nitroprusside 48-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-160 1458316-4 1992 Intravenous infusion of the vasodilating agent, nitroprusside, which lowered the blood pressure to levels comparable to that attained by hemorrhage, induced Fos-IR in greater than 65% of AVP-containing neurons in the SON, while relatively few AVP neurons in the PVN were Fos positive. Nitroprusside 48-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 271-274 1465218-1 1992 The effect of caerulein, an analog of cholecystokinin-8, on expression of the immediate-early genes c-fos and zif/268 was studied in the rat brain using Northern blot analysis and an in situ hybridization technique. Ceruletide 14-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 1465218-3 1992 Administration of the convulsant, pentylenetetrazole (PTZ), caused a dramatic increase of c-fos and zif/268 mRNAs in the hippocampus and dentate gyrus. Pentylenetetrazole 34-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 1465218-3 1992 Administration of the convulsant, pentylenetetrazole (PTZ), caused a dramatic increase of c-fos and zif/268 mRNAs in the hippocampus and dentate gyrus. Pentylenetetrazole 54-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 1465218-4 1992 Pretreatment with caerulein suppressed the PTZ-induced c-fos and zif/268 expression. Ceruletide 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 1465218-4 1992 Pretreatment with caerulein suppressed the PTZ-induced c-fos and zif/268 expression. Pentylenetetrazole 43-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 1360316-4 1992 Yohimbine, atipamezole and restraint stress each was found to cause increases in c-fos-like immunoreactivity (c-fos-li). Yohimbine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 1450906-1 1992 The effects of intraseptal application of atropine on c-fos proto-oncogene expression related to soman treatment were studied by immunohistochemistry for c-Fos-like proteins. Atropine 42-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 1450906-4 1992 The intraseptal application of atropine, which prevented soman-induced convulsions, reduced or even blocked c-Fos-like protein production related to soman treatment. Atropine 31-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 1450906-4 1992 The intraseptal application of atropine, which prevented soman-induced convulsions, reduced or even blocked c-Fos-like protein production related to soman treatment. Soman 149-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 1360316-4 1992 Yohimbine, atipamezole and restraint stress each was found to cause increases in c-fos-like immunoreactivity (c-fos-li). Yohimbine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 1360316-4 1992 Yohimbine, atipamezole and restraint stress each was found to cause increases in c-fos-like immunoreactivity (c-fos-li). atipamezole 11-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 1360316-4 1992 Yohimbine, atipamezole and restraint stress each was found to cause increases in c-fos-like immunoreactivity (c-fos-li). atipamezole 11-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 1360316-5 1992 Western blot analysis revealed increased c-fos protein in the cortex after yohimbine treatment. Yohimbine 75-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 1360316-6 1992 The c-fos-li response to yohimbine was blocked by prior administration of the beta receptor antagonist, dl-propranolol, and to a lesser degree by the alpha-1 antagonist, prazosin. Yohimbine 25-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 1360316-6 1992 The c-fos-li response to yohimbine was blocked by prior administration of the beta receptor antagonist, dl-propranolol, and to a lesser degree by the alpha-1 antagonist, prazosin. Propranolol 104-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 1360316-6 1992 The c-fos-li response to yohimbine was blocked by prior administration of the beta receptor antagonist, dl-propranolol, and to a lesser degree by the alpha-1 antagonist, prazosin. Prazosin 170-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 1362034-3 1992 The results pose an interesting problem, the possible relation of Fos protein to the biosynthesis of serotonin, awaiting further investigation. Serotonin 101-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 1359376-7 1992 C-fos and c-jun proto-oncogene activation was observed 1 h after reserpine in the locus ceruleus and adrenal medulla, specifically in those catecholaminergic structures that respond with increased enzyme gene transcription; in contrast, the dopaminergic neurons of the substantia nigra did not exhibit detectable proto-oncogene activation, only a small group of neurons in the ventral tegmental area showed c-fos without concomitant c-jun expression after reserpine. Reserpine 65-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 1328519-0 1992 Involvement of gamma-aminobutyric acid and N-methyl-D-aspartate receptors in the inhibitory effect of ethanol on pentylenetetrazole-induced c-fos expression in rat brain. gamma-Aminobutyric Acid 15-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 1328519-0 1992 Involvement of gamma-aminobutyric acid and N-methyl-D-aspartate receptors in the inhibitory effect of ethanol on pentylenetetrazole-induced c-fos expression in rat brain. Ethanol 102-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 1328519-0 1992 Involvement of gamma-aminobutyric acid and N-methyl-D-aspartate receptors in the inhibitory effect of ethanol on pentylenetetrazole-induced c-fos expression in rat brain. Pentylenetetrazole 113-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 1328519-1 1992 The expression of c-fos mRNA in rat brain was induced by intraperitoneal administration of pentylenetetrazole (PTZ) and picrotoxin, which act on the picrotoxin binding site of the gamma-aminobutyric acid-benzodiazepine (GABA-BZ) receptor complex, by N-methyl-D-aspartate (NMDA) and kainic acid, agonists of different classes of glutamate receptors and by caffeine, an antagonist of adenosine receptors. Pentylenetetrazole 91-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 1328519-1 1992 The expression of c-fos mRNA in rat brain was induced by intraperitoneal administration of pentylenetetrazole (PTZ) and picrotoxin, which act on the picrotoxin binding site of the gamma-aminobutyric acid-benzodiazepine (GABA-BZ) receptor complex, by N-methyl-D-aspartate (NMDA) and kainic acid, agonists of different classes of glutamate receptors and by caffeine, an antagonist of adenosine receptors. Pentylenetetrazole 111-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 1328519-1 1992 The expression of c-fos mRNA in rat brain was induced by intraperitoneal administration of pentylenetetrazole (PTZ) and picrotoxin, which act on the picrotoxin binding site of the gamma-aminobutyric acid-benzodiazepine (GABA-BZ) receptor complex, by N-methyl-D-aspartate (NMDA) and kainic acid, agonists of different classes of glutamate receptors and by caffeine, an antagonist of adenosine receptors. Picrotoxin 120-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 1328519-1 1992 The expression of c-fos mRNA in rat brain was induced by intraperitoneal administration of pentylenetetrazole (PTZ) and picrotoxin, which act on the picrotoxin binding site of the gamma-aminobutyric acid-benzodiazepine (GABA-BZ) receptor complex, by N-methyl-D-aspartate (NMDA) and kainic acid, agonists of different classes of glutamate receptors and by caffeine, an antagonist of adenosine receptors. Picrotoxin 149-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 1328519-1 1992 The expression of c-fos mRNA in rat brain was induced by intraperitoneal administration of pentylenetetrazole (PTZ) and picrotoxin, which act on the picrotoxin binding site of the gamma-aminobutyric acid-benzodiazepine (GABA-BZ) receptor complex, by N-methyl-D-aspartate (NMDA) and kainic acid, agonists of different classes of glutamate receptors and by caffeine, an antagonist of adenosine receptors. gamma-aminobutyric acid-benzodiazepine 180-218 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 1328519-1 1992 The expression of c-fos mRNA in rat brain was induced by intraperitoneal administration of pentylenetetrazole (PTZ) and picrotoxin, which act on the picrotoxin binding site of the gamma-aminobutyric acid-benzodiazepine (GABA-BZ) receptor complex, by N-methyl-D-aspartate (NMDA) and kainic acid, agonists of different classes of glutamate receptors and by caffeine, an antagonist of adenosine receptors. N-Methylaspartate 250-270 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 1328519-1 1992 The expression of c-fos mRNA in rat brain was induced by intraperitoneal administration of pentylenetetrazole (PTZ) and picrotoxin, which act on the picrotoxin binding site of the gamma-aminobutyric acid-benzodiazepine (GABA-BZ) receptor complex, by N-methyl-D-aspartate (NMDA) and kainic acid, agonists of different classes of glutamate receptors and by caffeine, an antagonist of adenosine receptors. N-Methylaspartate 272-276 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 1328519-1 1992 The expression of c-fos mRNA in rat brain was induced by intraperitoneal administration of pentylenetetrazole (PTZ) and picrotoxin, which act on the picrotoxin binding site of the gamma-aminobutyric acid-benzodiazepine (GABA-BZ) receptor complex, by N-methyl-D-aspartate (NMDA) and kainic acid, agonists of different classes of glutamate receptors and by caffeine, an antagonist of adenosine receptors. Kainic Acid 282-293 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 1328519-1 1992 The expression of c-fos mRNA in rat brain was induced by intraperitoneal administration of pentylenetetrazole (PTZ) and picrotoxin, which act on the picrotoxin binding site of the gamma-aminobutyric acid-benzodiazepine (GABA-BZ) receptor complex, by N-methyl-D-aspartate (NMDA) and kainic acid, agonists of different classes of glutamate receptors and by caffeine, an antagonist of adenosine receptors. Caffeine 355-363 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 1328519-3 1992 Ro 15-4513 partially reversed the inhibitory effect of ethanol on PTZ-induced c-fos mRNA synthesis. Ro 15-4513 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 1328519-3 1992 Ro 15-4513 partially reversed the inhibitory effect of ethanol on PTZ-induced c-fos mRNA synthesis. Ethanol 55-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 1328519-3 1992 Ro 15-4513 partially reversed the inhibitory effect of ethanol on PTZ-induced c-fos mRNA synthesis. Pentylenetetrazole 66-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 1328519-5 1992 Taken together, these data suggest that ethanol blocks c-fos gene activation by noncompetitive antagonists of the GABA-BZ receptor via actions on both the NMDA and GABA receptors. Ethanol 40-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 1333540-7 1992 In early passage cells, treatment with the beta-adrenergic agonist, isoproterenol (IPR), resulted in an increase in c-fos mRNA and a decrease in c-jun mRNA (Gu-bits RM, Yu H: J Neurosci Res, 30:625-630, 1991). Isoproterenol 68-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 1357113-0 1992 D1-like and D2-like dopamine receptors synergistically activate rotation and c-fos expression in the dopamine-depleted striatum in a rat model of Parkinson"s disease. Dopamine 20-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 1357113-0 1992 D1-like and D2-like dopamine receptors synergistically activate rotation and c-fos expression in the dopamine-depleted striatum in a rat model of Parkinson"s disease. Dopamine 101-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 1357113-4 1992 In rats with unilateral 6-hydroxydopamine (6-OHDA) lesions of the nigrostriatal pathway, D1-selective (but not D2-selective) dopamine agonists produce a marked increase in expression of the immediate-early gene c-fos in the striatum ipsilateral to the 6-OHDA lesion. Oxidopamine 24-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 211-216 1357113-4 1992 In rats with unilateral 6-hydroxydopamine (6-OHDA) lesions of the nigrostriatal pathway, D1-selective (but not D2-selective) dopamine agonists produce a marked increase in expression of the immediate-early gene c-fos in the striatum ipsilateral to the 6-OHDA lesion. Oxidopamine 43-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 211-216 1357113-6 1992 We examined the effects of the D1-selective agonist SKF-38393 and the D2-selective agonist quinpirole (LY 171555) on the expression of Fos-like protein and c-fos mRNA in the caudoputamen and made parallel behavioral observations in the same animals. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 52-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-138 1357113-6 1992 We examined the effects of the D1-selective agonist SKF-38393 and the D2-selective agonist quinpirole (LY 171555) on the expression of Fos-like protein and c-fos mRNA in the caudoputamen and made parallel behavioral observations in the same animals. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 52-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 1357113-6 1992 We examined the effects of the D1-selective agonist SKF-38393 and the D2-selective agonist quinpirole (LY 171555) on the expression of Fos-like protein and c-fos mRNA in the caudoputamen and made parallel behavioral observations in the same animals. Quinpirole 91-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-138 1357113-6 1992 We examined the effects of the D1-selective agonist SKF-38393 and the D2-selective agonist quinpirole (LY 171555) on the expression of Fos-like protein and c-fos mRNA in the caudoputamen and made parallel behavioral observations in the same animals. Quinpirole 91-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 1357113-6 1992 We examined the effects of the D1-selective agonist SKF-38393 and the D2-selective agonist quinpirole (LY 171555) on the expression of Fos-like protein and c-fos mRNA in the caudoputamen and made parallel behavioral observations in the same animals. 4,4a,5,6,7,8,8a,9-octahydro-5-propyl-1H-pyrzolo(3,4-g)quinoline 103-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-138 1357113-6 1992 We examined the effects of the D1-selective agonist SKF-38393 and the D2-selective agonist quinpirole (LY 171555) on the expression of Fos-like protein and c-fos mRNA in the caudoputamen and made parallel behavioral observations in the same animals. 4,4a,5,6,7,8,8a,9-octahydro-5-propyl-1H-pyrzolo(3,4-g)quinoline 103-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 1357113-8 1992 A low dose of quinpirole elicited contralateral rotation but little or no induction of Fos-like immunoreactivity in the caudoputamen; there was, however, induction of Fos in the globus pallidus ipsilateral to the 6-OHDA lesion. Quinpirole 14-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 167-170 1357113-9 1992 Combination of the low dose of SKF-38393 and quinpirole produced a synergistic effect on rotation and elicited, in the dopamine-depleted caudoputamen, a striking pattern of Fos-like protein expression in which Fos-positive neurons were concentrated in striosomes and in the dorsolateral caudoputamen. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 31-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 173-176 1357113-9 1992 Combination of the low dose of SKF-38393 and quinpirole produced a synergistic effect on rotation and elicited, in the dopamine-depleted caudoputamen, a striking pattern of Fos-like protein expression in which Fos-positive neurons were concentrated in striosomes and in the dorsolateral caudoputamen. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 31-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 210-213 1357113-9 1992 Combination of the low dose of SKF-38393 and quinpirole produced a synergistic effect on rotation and elicited, in the dopamine-depleted caudoputamen, a striking pattern of Fos-like protein expression in which Fos-positive neurons were concentrated in striosomes and in the dorsolateral caudoputamen. Quinpirole 45-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 173-176 1357113-9 1992 Combination of the low dose of SKF-38393 and quinpirole produced a synergistic effect on rotation and elicited, in the dopamine-depleted caudoputamen, a striking pattern of Fos-like protein expression in which Fos-positive neurons were concentrated in striosomes and in the dorsolateral caudoputamen. Quinpirole 45-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 210-213 1357113-9 1992 Combination of the low dose of SKF-38393 and quinpirole produced a synergistic effect on rotation and elicited, in the dopamine-depleted caudoputamen, a striking pattern of Fos-like protein expression in which Fos-positive neurons were concentrated in striosomes and in the dorsolateral caudoputamen. Dopamine 119-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 173-176 1357113-9 1992 Combination of the low dose of SKF-38393 and quinpirole produced a synergistic effect on rotation and elicited, in the dopamine-depleted caudoputamen, a striking pattern of Fos-like protein expression in which Fos-positive neurons were concentrated in striosomes and in the dorsolateral caudoputamen. Dopamine 119-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 210-213 1357113-11 1992 Both the contraversive rotation and the induction of Fos-like immunoreactivity were blocked by the preadministration of the D1-preferring antagonist SCH-23390 and the D2-selective antagonist raclopride in combination. SCH 23390 149-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 1357113-11 1992 Both the contraversive rotation and the induction of Fos-like immunoreactivity were blocked by the preadministration of the D1-preferring antagonist SCH-23390 and the D2-selective antagonist raclopride in combination. Raclopride 191-201 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 1357113-12 1992 Pretreatment with the glutamate NMDA receptor antagonist MK-801 also blocked the induction of Fos-like immunoreactivity, and it reversed the rotation. Dizocilpine Maleate 57-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 1403102-3 1992 To understand better the neurobiology of cocaine-induced environment-specific conditioning, Fos expression was examined in the forebrain of rats exposed to an environment in which they had previously received cocaine. Cocaine 41-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 1403102-5 1992 Consistent with its stimulant actions, cocaine produced an increase in locomotion that was accompanied by an increase in Fos expression within specific limbic regions (cingulate cortex, claustrum, piriform cortex, lateral septal nucleus, paraventricular nucleus of the thalamus, lateral habenula, and amygdala) as well as the basal ganglia (dorsomedial striatum and nucleus accumbens). Cocaine 39-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-124 1421082-1 1992 Detection of Fos protein expression with a polyclonal antibody was used to identify brainstem neurons responding to acute (24 h) effects of a unilateral sodium arsanilate chemical labyrinthectomy in Long-Evans rats. Arsanilic Acid 153-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 1421085-0 1992 Nitric oxide mediates Fos expression in the spinal cord induced by mechanical noxious stimulation. Nitric Oxide 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-25 1421085-1 1992 Immunocytochemical localization of Fos protein was used to analyze the involvement of nitric oxide (NO) in the expression of Fos in the spinal cord, induced by mechanical noxious stimulation (NS). Nitric Oxide 86-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 1421085-1 1992 Immunocytochemical localization of Fos protein was used to analyze the involvement of nitric oxide (NO) in the expression of Fos in the spinal cord, induced by mechanical noxious stimulation (NS). Nitric Oxide 86-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-128 1421085-3 1992 Pretreatment with L-NAME but not its stereoisomer N omega-nitro-D-arginine methyl ester (D-NAME), produced a dose-dependent suppression of Fos expression induced by mechanical noxious stimulation. NG-Nitroarginine Methyl Ester 18-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 1421086-1 1992 The present paper describes the effect of capsaicin-induced stressful stimulus on the expression of immediate early genes (IEGs) c-fos, c-jun, junB and junD in the hypothalamic paraventricular nucleus (PVN) and the central amygdaloid nucleus (ACe) using in situ hybridization. Capsaicin 42-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 1436507-3 1992 Males bearing unilateral electrothermal lesions of the olfactory peduncle showed a significant reduction in c-fos expression in the ipsilateral medial amygdala, but not in other structures, provided their coital interaction with oestrous females was restricted to mount-thrust and occasional intromissive patterns due to repeated application of lidocaine anaesthetic to the penis. Lidocaine 345-354 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 1436508-0 1992 Narine occlusion decreases basal levels of Fos protein in the cerebral cortex of the lizard Podarcis hispanica. narine 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 1448203-3 1992 After acute and chronic dehydration by salt-loading, the number of Fos-like positive neurons increased dramatically. Salts 39-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 1384044-3 1992 Cocaine injected into pregnant rats or directly into rat fetuses on day 20 of gestation selectively activated c-fos gene expression in the fetal SCN; cocaine did not induce c-fos expression elsewhere in the fetal brain or in the maternal SCN. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 1384044-4 1992 This cocaine-induced activation of c-fos expression in fetal SCN was mediated in part through D1-dopamine receptors, as the cocaine-induced activation was partially blocked by the D1-dopamine receptor antagonist SCH 23390. Cocaine 5-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 1384044-4 1992 This cocaine-induced activation of c-fos expression in fetal SCN was mediated in part through D1-dopamine receptors, as the cocaine-induced activation was partially blocked by the D1-dopamine receptor antagonist SCH 23390. Cocaine 124-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 1384044-5 1992 In addition, the selective D1-dopamine receptor agonist SKF 38393 induced high levels of c-fos expression in the fetal SCN. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 56-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 1520705-2 1992 PMA, PDB and O-met-PMA, but not alpha-phorbol, stimulated c-fos expression. o-met-pma 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 1520705-7 1992 Actinomycin D markedly diminished basal c-fos expression whereas cycloheximide had the opposite effect. Dactinomycin 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 1520705-9 1992 PMA and the calcium-mobilizing hormones increased c-fos expression above the level observed with cycloheximide alone. Calcium 12-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 1520705-9 1992 PMA and the calcium-mobilizing hormones increased c-fos expression above the level observed with cycloheximide alone. Cycloheximide 97-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 1520705-10 1992 These data suggest that PMA and the calcium-mobilizing hormones increased both transcription of the c-fos gene and stabilization of the proto-oncogene mRNA. Calcium 36-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 1415544-0 1992 CCK, bombesin, and carbachol stimulate c-fos, c-jun, and c-myc oncogene expression in rat pancreatic acini. Carbachol 19-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 1511096-6 1992 The protein kinase inhibitor, 2-aminopurine (2-AP), inhibited the FSH/IN-induced increases in c-myc and c-fos mRNA levels, the percentage of cells staining for Myc and Fos protein, and DNA and protein synthesis. 2-Aminopurine 30-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 1511096-6 1992 The protein kinase inhibitor, 2-aminopurine (2-AP), inhibited the FSH/IN-induced increases in c-myc and c-fos mRNA levels, the percentage of cells staining for Myc and Fos protein, and DNA and protein synthesis. 2-Aminopurine 30-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-171 1511096-6 1992 The protein kinase inhibitor, 2-aminopurine (2-AP), inhibited the FSH/IN-induced increases in c-myc and c-fos mRNA levels, the percentage of cells staining for Myc and Fos protein, and DNA and protein synthesis. 2-Aminopurine 45-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 1511096-6 1992 The protein kinase inhibitor, 2-aminopurine (2-AP), inhibited the FSH/IN-induced increases in c-myc and c-fos mRNA levels, the percentage of cells staining for Myc and Fos protein, and DNA and protein synthesis. 2-Aminopurine 45-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-171 1356602-2 1992 A single injection IP of haloperidol (2 mg/kg) or sulpiride (100 mg/kg) produced a rapid and transient increase in c-jun, zif-268 and c-fos mRNA. Haloperidol 25-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-139 1356602-2 1992 A single injection IP of haloperidol (2 mg/kg) or sulpiride (100 mg/kg) produced a rapid and transient increase in c-jun, zif-268 and c-fos mRNA. Sulpiride 50-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-139 1527597-1 1992 The expression of Fos, the protein product of the primary response gene c-fos, was used metabolically to map the short-term (1 hr) effects of urethane and sodium pentobarbital anesthesia in rat. Urethane 142-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 1322970-4 1992 The D1 antagonist SCH-23390 blocked GBR-12909-induced activation of Fos while potentiating the stimulation of DA output. SCH 23390 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 1527597-1 1992 The expression of Fos, the protein product of the primary response gene c-fos, was used metabolically to map the short-term (1 hr) effects of urethane and sodium pentobarbital anesthesia in rat. Urethane 142-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 1527597-1 1992 The expression of Fos, the protein product of the primary response gene c-fos, was used metabolically to map the short-term (1 hr) effects of urethane and sodium pentobarbital anesthesia in rat. Pentobarbital 155-175 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 1527597-1 1992 The expression of Fos, the protein product of the primary response gene c-fos, was used metabolically to map the short-term (1 hr) effects of urethane and sodium pentobarbital anesthesia in rat. Pentobarbital 155-175 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 1407557-1 1992 Immunocytochemical technique was used to study the distribution of c-FOS protein immunoreactive cells in the spinal cord and gracile nuclei 2 h after electrical stimulation of the sciatic nerve in ketamine/xylazine/acepromazine-anesthetized adult rats. Ketamine 197-205 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 1407557-1 1992 Immunocytochemical technique was used to study the distribution of c-FOS protein immunoreactive cells in the spinal cord and gracile nuclei 2 h after electrical stimulation of the sciatic nerve in ketamine/xylazine/acepromazine-anesthetized adult rats. Xylazine 206-214 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 1407557-1 1992 Immunocytochemical technique was used to study the distribution of c-FOS protein immunoreactive cells in the spinal cord and gracile nuclei 2 h after electrical stimulation of the sciatic nerve in ketamine/xylazine/acepromazine-anesthetized adult rats. Acepromazine 215-227 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 1292248-0 1992 Expressions of c-fos and c-myc genes during 3"-methyl-4-dimethylaminoazobenzene (3"-MeDAB)-induced rat hepatocarcinoma. Methyldimethylaminoazobenzene 44-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 1359024-0 1992 Dopamine and glutamate agonists stimulate neuron-specific expression of Fos-like protein in the striatum. Dopamine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 1359024-0 1992 Dopamine and glutamate agonists stimulate neuron-specific expression of Fos-like protein in the striatum. Glutamic Acid 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 1359024-3 1992 To study the effects of these neurotransmitters on gene transcription in striatal neurons, we treated rats with dopamine (monoamine) agonists and with glutamate agonists and monitored the induction of Fos-like protein in striatal neurons. Glutamic Acid 151-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 201-204 1359024-7 1992 Both the indirect monoamine agonists and the glutamate receptor agonist stimulated rapid nuclear expression of Fos-like protein in specific classes of striatal neurons. monoamine 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 1359024-10 1992 The monoamine and glutamate agonists both induced Fos-like protein exclusively in striatal neurons that constitutively expressed the protein phosphatase inhibitor DARPP-32 (dopamine and cAMP-regulated phosphoprotein). monoamine 4-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 1359024-10 1992 The monoamine and glutamate agonists both induced Fos-like protein exclusively in striatal neurons that constitutively expressed the protein phosphatase inhibitor DARPP-32 (dopamine and cAMP-regulated phosphoprotein). Glutamic Acid 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 1359024-10 1992 The monoamine and glutamate agonists both induced Fos-like protein exclusively in striatal neurons that constitutively expressed the protein phosphatase inhibitor DARPP-32 (dopamine and cAMP-regulated phosphoprotein). Dopamine 173-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 1359024-10 1992 The monoamine and glutamate agonists both induced Fos-like protein exclusively in striatal neurons that constitutively expressed the protein phosphatase inhibitor DARPP-32 (dopamine and cAMP-regulated phosphoprotein). Cyclic AMP 186-190 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 1359024-15 1992 Glutamate agonist stimulation, but not dopamine agonist stimulation, induced Fos-like protein in a subpopulation of striatal interneurons, namely, a group of neurons exhibiting NADPH-diaphorase activity. Glutamic Acid 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-80 1353702-2 1992 We had previously described the paradox of finding similar cascades of gene activity (c-fos greater than c-myc greater than hsp-70) induced during the early period of ventral prostate regression and during the regrowth of the ventral prostate gland initiated by testosterone replenishment. Testosterone 262-274 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 1517486-1 1992 Water deprivation induces the production of the transcription factor Fos in neurons of the neurohypophysial system. Water 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 1517486-6 1992 Faint nuclear Fos immunostaining was found in the organum vasculosum of the lamina terminalis (OVLT), the median preoptic nucleus (MnPO), subfornical organ (SFO), and SON of non-injected and isotonic saline-injected Brattleboro but not Long-Evans rats. Sodium Chloride 200-206 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 1517486-7 1992 Hypertonic saline injection specifically induced Fos-IR in neurons located in the SFO, OVLT, MnPO, PVN, SON, hypothalamic accessory nuclei (including the nucleus circularis), and arcuate hypothalamic nucleus (Arc) in both Long Evans and Brattleboro rats. Sodium Chloride 11-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 1517486-9 1992 Stress of handling and (isotonic saline) injection induced Fos-IR in the lateral septal nuclei, central amygdaloid nuclei, medial amygdaloid nucleus, medial preoptic area, the bed nucleus of the stria terminalis, cingulate- and piriform cortex, the lateral hypothalamic area, ventromedial hypothalamic nucleus, and the habenular nucleus. Sodium Chloride 33-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 1393570-3 1992 with hypertonic saline expressed c-fos-like immunoreactivity (FLI) in the paraventricular (PVN), periventricular (PEV) and supraoptic (SON) hypothalamic nuclei, and in the preoptic and retrochiasmatic regions, as early as 30 min after stimulation and up to 6 h, while these areas were mostly devoid of staining in isotonic saline-injected animals. Sodium Chloride 16-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 1393570-3 1992 with hypertonic saline expressed c-fos-like immunoreactivity (FLI) in the paraventricular (PVN), periventricular (PEV) and supraoptic (SON) hypothalamic nuclei, and in the preoptic and retrochiasmatic regions, as early as 30 min after stimulation and up to 6 h, while these areas were mostly devoid of staining in isotonic saline-injected animals. Sodium Chloride 323-329 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 1397060-10 1992 ), expression of c-fos mRNA was observed in the hypothalamus and in brain areas involved in sensory processing; these effects were totally blocked by pretreatment with 2 mg/kg scopolamine. Scopolamine 176-187 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 1573410-1 1992 Administration of kainate or pentylenetetrazole increased c-fos, c-jun, junB, and junD mRNA levels in rat brain in a dose-dependent manner. Kainic Acid 18-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 1292248-0 1992 Expressions of c-fos and c-myc genes during 3"-methyl-4-dimethylaminoazobenzene (3"-MeDAB)-induced rat hepatocarcinoma. Methyldimethylaminoazobenzene 81-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 1292248-8 1992 We have found that the rate of [3H] thymidine incorporation corresponds to the elevated levels of c-fos and c-myc transcripts in the precancerous stages. Tritium 32-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 1292248-8 1992 We have found that the rate of [3H] thymidine incorporation corresponds to the elevated levels of c-fos and c-myc transcripts in the precancerous stages. Thymidine 36-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 1642355-0 1992 Selective effects of pentobarbital and halothane on c-fos and jun-B gene expression in rat brain. Pentobarbital 21-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 1642355-0 1992 Selective effects of pentobarbital and halothane on c-fos and jun-B gene expression in rat brain. Halothane 39-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 1642355-1 1992 The effects of pentobarbital and halothane anesthesia on the expression in brain of the immediate-early genes c-fos and jun-B were investigated. Pentobarbital 15-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 1642355-1 1992 The effects of pentobarbital and halothane anesthesia on the expression in brain of the immediate-early genes c-fos and jun-B were investigated. Halothane 33-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 1280214-0 1992 ACTH increases expression of c-fos, c-jun and beta-actin genes in the dexamethasone-treated rat adrenals. Dexamethasone 70-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 1280214-6 1992 It was demonstrated that ACTH increases the mRNAs coding c-fos and c-jun in the adrenal glands of dexamethasone-treated, ACTH-suppressed rats. Dexamethasone 98-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 1354257-1 1992 Administration of the selective D1-dopamine receptor agonist 2,3,4,5-tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine (SKF-38393) to neonatal 6-hydroxydopamine-lesioned rats results in profound behavioral manifestations and induction of striatal c-fos-like immunoreactivity. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 61-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 248-253 1354257-2 1992 The full D1-dopamine agonist I,[R,S]1-aminomethyl-3,4-dihydro-5,6-dihydroxy-3-phenyl-1H-2-benzopyran hydrochloride (A-68930), like SKF-38393, produced a dose-dependent, D1-selective increase in locomotor activity and striatal c-fos-like immunoreactivity. [r,s]1-aminomethyl-3,4-dihydro-5,6-dihydroxy-3-phenyl-1h-2-benzopyran hydrochloride 31-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 226-231 1501769-1 1992 The expression of c-fos protein was examined by immunohistochemistry in serial sections of brainstem following the instillation of either autologous arterial blood (0.3 ml) or mock cerebrospinal fluid (0.3 ml) through a catheter placed in the cisterna magna, or following catheter placement alone in pentobarbital-anesthetized Sprague-Dawley rats. Pentobarbital 300-313 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 1293753-6 1992 The c-fos expression was very low in control animals receiving neither formalin nor EA administration. Formaldehyde 71-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 1506406-5 1992 Thirteen to 15% CO2 evoked fos-like immunoreactivity (FLI) in 321 +/- 146 neurons/rat. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 16-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 1506406-9 1992 We suggest that respiratory CO2 chemoreceptor neurons can be identified in rats by their expression of c-fos after 1 h of hypercapnia. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 28-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 1613551-10 1992 These patterns of NGFI-A activation are remarkably similar to those found for Fos-like immunoreactivity following acute amphetamine and cocaine treatments, suggesting that coordinate activation of members of at least two immediate-early gene families occurs in the striatum following catecholaminergic stimulation. Amphetamine 120-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 1613551-10 1992 These patterns of NGFI-A activation are remarkably similar to those found for Fos-like immunoreactivity following acute amphetamine and cocaine treatments, suggesting that coordinate activation of members of at least two immediate-early gene families occurs in the striatum following catecholaminergic stimulation. Cocaine 136-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 1324419-9 1992 Additional sections from metyrapone-and vehicle-treated rats were hybridized with probes complementary to mRNA encoding the immediate-early gene c-fos. Metyrapone 25-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-150 1324419-10 1992 c-fos was not present under unstimulated conditions yet was rapidly induced upon metyrapone treatment or vehicle injection (15 min). Metyrapone 81-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 1631058-6 1992 Similarly, levels of Fos-like immunoreactivity, which are increased in the NAc by acute cocaine, were reduced to control levels in chronic cocaine-treated rats. Cocaine 88-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 1631058-6 1992 Similarly, levels of Fos-like immunoreactivity, which are increased in the NAc by acute cocaine, were reduced to control levels in chronic cocaine-treated rats. Cocaine 139-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 1631058-9 1992 In contrast, AP-1 binding activity in the NAc of animals treated chronically with cocaine remained elevated at acute levels 18 hr after the last chronic injection, a time at which c-fos and c-jun mRNA levels and Fos-like immunoreactivity had returned to control values. Cocaine 82-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 180-185 1631058-9 1992 In contrast, AP-1 binding activity in the NAc of animals treated chronically with cocaine remained elevated at acute levels 18 hr after the last chronic injection, a time at which c-fos and c-jun mRNA levels and Fos-like immunoreactivity had returned to control values. Cocaine 82-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 212-215 1498673-1 1992 In situ hybridization for c-fos mRNA was performed on brain sections (a) from rats after an acute cocaine-induced seizure or from saline-injected controls and (b) from rats after their first cocaine-kindled seizure, as well as from rats that had not yet developed cocaine-kindled seizures (but were exposed to the same amount of cocaine as those that did exhibit convulsions) and from saline-injected controls. Cocaine 98-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 1498673-1 1992 In situ hybridization for c-fos mRNA was performed on brain sections (a) from rats after an acute cocaine-induced seizure or from saline-injected controls and (b) from rats after their first cocaine-kindled seizure, as well as from rats that had not yet developed cocaine-kindled seizures (but were exposed to the same amount of cocaine as those that did exhibit convulsions) and from saline-injected controls. Sodium Chloride 130-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 1498673-1 1992 In situ hybridization for c-fos mRNA was performed on brain sections (a) from rats after an acute cocaine-induced seizure or from saline-injected controls and (b) from rats after their first cocaine-kindled seizure, as well as from rats that had not yet developed cocaine-kindled seizures (but were exposed to the same amount of cocaine as those that did exhibit convulsions) and from saline-injected controls. Cocaine 191-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 1498673-1 1992 In situ hybridization for c-fos mRNA was performed on brain sections (a) from rats after an acute cocaine-induced seizure or from saline-injected controls and (b) from rats after their first cocaine-kindled seizure, as well as from rats that had not yet developed cocaine-kindled seizures (but were exposed to the same amount of cocaine as those that did exhibit convulsions) and from saline-injected controls. Cocaine 191-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 1498673-1 1992 In situ hybridization for c-fos mRNA was performed on brain sections (a) from rats after an acute cocaine-induced seizure or from saline-injected controls and (b) from rats after their first cocaine-kindled seizure, as well as from rats that had not yet developed cocaine-kindled seizures (but were exposed to the same amount of cocaine as those that did exhibit convulsions) and from saline-injected controls. Sodium Chloride 385-391 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 1498673-2 1992 Increased expression of c-fos mRNA was observed in animals demonstrating cocaine-induced seizures acutely or following pharmacological kindling. Cocaine 73-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 1498673-3 1992 Rats that experienced acute seizures after cocaine (65 mg/kg) showed pronounced increase in expression of c-fos mRNA in the dentate gyrus of the hippocampus and olfactory bulb. Cocaine 43-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 1498673-5 1992 In rats that were injected daily with an initially subconvulsive dose of cocaine-HCl (40 mg/kg), the cocaine-kindled seizures induced elevations in c-fos mRNA in the same brain regions as with an acute cocaine-induced seizure with the single exception of the VMH. Cocaine 73-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 1498673-5 1992 In rats that were injected daily with an initially subconvulsive dose of cocaine-HCl (40 mg/kg), the cocaine-kindled seizures induced elevations in c-fos mRNA in the same brain regions as with an acute cocaine-induced seizure with the single exception of the VMH. Cocaine 73-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 1498673-6 1992 These findings not only suggest the involvement of limbic, cortical and striatal structures in the cocaine-induced seizure, but also raise the possibility that alterations in the proto-oncogene c-fos and its subsequent impact on gene expression could play a role in the changes in neural excitability associated with cocaine-induced kindling. Cocaine 99-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-199 1498673-6 1992 These findings not only suggest the involvement of limbic, cortical and striatal structures in the cocaine-induced seizure, but also raise the possibility that alterations in the proto-oncogene c-fos and its subsequent impact on gene expression could play a role in the changes in neural excitability associated with cocaine-induced kindling. Cocaine 317-324 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-199 1327382-0 1992 CP-93,129, sumatriptan, dihydroergotamine block c-fos expression within rat trigeminal nucleus caudalis caused by chemical stimulation of the meninges. 2-Ethyl-1-methylpyridin-4-one 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 1327382-0 1992 CP-93,129, sumatriptan, dihydroergotamine block c-fos expression within rat trigeminal nucleus caudalis caused by chemical stimulation of the meninges. Sumatriptan 11-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 1327382-0 1992 CP-93,129, sumatriptan, dihydroergotamine block c-fos expression within rat trigeminal nucleus caudalis caused by chemical stimulation of the meninges. Dihydroergotamine 24-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 1327382-3 1992 C-fos was induced by injecting an algesic, vasoconstrictor substance (0.3 ml of autologous blood) or a pro-inflammatory molecule, carrageenin (1 mg in 0.1 ml saline) into the cisterna magna of pentobarbitone-anaesthetized Sprague-Dawley rats and was visualized in serial sections (50 micrometers) by use of a polyclonal antiserum. Carrageenan 130-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 1327382-3 1992 C-fos was induced by injecting an algesic, vasoconstrictor substance (0.3 ml of autologous blood) or a pro-inflammatory molecule, carrageenin (1 mg in 0.1 ml saline) into the cisterna magna of pentobarbitone-anaesthetized Sprague-Dawley rats and was visualized in serial sections (50 micrometers) by use of a polyclonal antiserum. Sodium Chloride 158-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 1327382-3 1992 C-fos was induced by injecting an algesic, vasoconstrictor substance (0.3 ml of autologous blood) or a pro-inflammatory molecule, carrageenin (1 mg in 0.1 ml saline) into the cisterna magna of pentobarbitone-anaesthetized Sprague-Dawley rats and was visualized in serial sections (50 micrometers) by use of a polyclonal antiserum. Pentobarbital 193-207 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 1327382-16 1992 reduced the number of c-fos labelled cells by 47% within lamina I, IIo after carrageenin instillation. Carrageenan 77-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 8475076-8 1993 Administration of the noncompetitive N-methyl-D-aspartate receptor antagonist MK801 did not attenuate the odor induction of c-fos but, instead, increased c-fos mRNA levels throughout the bulb. Dizocilpine Maleate 78-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 1511346-0 1992 c-fos expression in noradrenergic A2 neurons of the rat during the estrous cycle and after steroid hormone treatments. Steroids 91-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 1504828-3 1992 Amphetamine (5 mg/kg; 2 h) induced Fos-like immunoreactivity in clusters of cells located mainly within the DARPP-32-positive areas within the transplants, i.e. within the striatum-like graft compartment which is preferentially innervated by the host DA afferents. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 1504828-6 1992 Two to 3 weeks after a 6-OHDA lesion of the host DA pathway (i.e. a time sufficient for DA receptor supersensitivity to develop), apomorphine-induced extensive Fos-activation selectively within the DARPP-32-positive areas of the graft. Apomorphine 130-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-163 1504828-9 1992 In intrastriatal grafts of fetal neocortical tissue, which were studied for comparison, the amphetamine- and apomorphine-induced effects on Fos expression were much smaller and similar to that seen in the DARPP-32-negative, non-striatal compartment within the striatal grafts. Amphetamine 92-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-143 1504828-9 1992 In intrastriatal grafts of fetal neocortical tissue, which were studied for comparison, the amphetamine- and apomorphine-induced effects on Fos expression were much smaller and similar to that seen in the DARPP-32-negative, non-striatal compartment within the striatal grafts. Apomorphine 109-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-143 1317778-0 1992 A 3",5"-cyclic adenosine monophosphate-dependent pathway is responsible for a rapid increase in c-fos messenger ribonucleic acid by adrenocorticotropin. 3",5"-cyclic adenosine monophosphate 2-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 1375896-0 1992 Progesterone inhibits the estrogen-induced expression of c-fos messenger ribonucleic acid in the uterus. Progesterone 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 1375896-1 1992 Estradiol produces a large increase in the uterine level of c-fos mRNA, which is maximum in 3 h. The administration of progesterone antagonizes this estrogen-induced increase in protooncogene transcript levels in both the rat and mouse. Estradiol 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 1375896-1 1992 Estradiol produces a large increase in the uterine level of c-fos mRNA, which is maximum in 3 h. The administration of progesterone antagonizes this estrogen-induced increase in protooncogene transcript levels in both the rat and mouse. Progesterone 119-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 1375896-2 1992 The inhibitory effect of progesterone is observed within 1 h after hormone treatment and persists for 9-18 h. In the rat, this effect can be observed at a dose of 0.25 mg progesterone and is maximum at a dose of 2.5 mg. A similar inhibition of fos mRNA levels after estrogen administration is produced by the glucocorticoid dexamethasone, but not by androgens or mineralocorticoids. Progesterone 25-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 244-247 1375896-4 1992 Uterine levels of c-fos mRNA observed after treatment with the phorbol ester phorbol 12-myristate 13-acetate are not decreased by a 3-h pretreatment with progesterone. Phorbol Esters 63-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 1375896-4 1992 Uterine levels of c-fos mRNA observed after treatment with the phorbol ester phorbol 12-myristate 13-acetate are not decreased by a 3-h pretreatment with progesterone. Tetradecanoylphorbol Acetate 77-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 1383560-0 1992 AP-1 complex and c-fos transcription are involved in TPA provoked and trans-synaptic inductions of the tyrosine hydroxylase gene: insights into long-term regulatory mechanisms. Tetradecanoylphorbol Acetate 53-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 1573410-1 1992 Administration of kainate or pentylenetetrazole increased c-fos, c-jun, junB, and junD mRNA levels in rat brain in a dose-dependent manner. Pentylenetetrazole 29-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 1573410-4 1992 Adrenalectomy significantly potentiated kainate-induced increases, compared with increases caused by kainate (4 mg/kg) alone, in the hippocampal mRNA levels of c-fos and junB by 6.5-fold and of junD by twofold and tended to augment c-jun mRNA. Kainic Acid 40-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 1573410-4 1992 Adrenalectomy significantly potentiated kainate-induced increases, compared with increases caused by kainate (4 mg/kg) alone, in the hippocampal mRNA levels of c-fos and junB by 6.5-fold and of junD by twofold and tended to augment c-jun mRNA. Kainic Acid 101-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 1573410-6 1992 Adrenalectomy also significantly increased pentylenetetrazole-induced levels of c-fos mRNA in the cortex. Pentylenetetrazole 43-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 1391749-3 1992 H2O2 also increases c-fos mRNA expression, which is probably involved in the gene regulation of both NGF and bFGF. Hydrogen Peroxide 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 1327843-0 1992 Expression of Fos-like immunoreactivity by yohimbine and clonidine in the rat brain. Yohimbine 43-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 19912866-5 1992 Lactation inhibited hippocampal and cortical cFos induction in response to NMA, which was consistent with the absence of behavioral responses. nma 75-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-49 19912866-8 1992 In contrast, treatment with kainate (an agonist for a different type of glutamate receptor) induced similar patterns of cFos expression and behavioral responses (wet-dog shakes) in cycling and lactating rats. Kainic Acid 28-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-124 1354554-0 1992 The effect of glutamate antagonists on c-fos expression induced in spinal neurons by irritation of the lower urinary tract. Glutamic Acid 14-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 1327843-0 1992 Expression of Fos-like immunoreactivity by yohimbine and clonidine in the rat brain. Clonidine 57-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 1374894-0 1992 Differential expression of c-fos and zif268 in rat striatum after haloperidol, clozapine, and amphetamine. Haloperidol 66-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 1327843-5 1992 suppressed the Fos expression by yohimbine in these nuclei, and clonidine (100 micrograms/kg, i.p.) Yohimbine 33-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-18 1374894-0 1992 Differential expression of c-fos and zif268 in rat striatum after haloperidol, clozapine, and amphetamine. Clozapine 79-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 1374894-0 1992 Differential expression of c-fos and zif268 in rat striatum after haloperidol, clozapine, and amphetamine. Amphetamine 94-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 1407673-3 1992 with hypertonic saline solution as early as 30 min after stimulation, and the effect lasted up to 3 h. Only a few c-Fos-positive cells were detected in the SFO of rats injected with isotonic saline. Sodium Chloride 191-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 1374894-6 1992 Antipsychotic drugs with potent dopamine D2 receptor antagonist properties, such as haloperidol, induced both c-fos and zif268 mRNA in the caudate-putamen; however, the atypical antipsychotic drug clozapine induced zif268 but not c-fos mRNA in that region. Haloperidol 84-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 1374894-6 1992 Antipsychotic drugs with potent dopamine D2 receptor antagonist properties, such as haloperidol, induced both c-fos and zif268 mRNA in the caudate-putamen; however, the atypical antipsychotic drug clozapine induced zif268 but not c-fos mRNA in that region. Haloperidol 84-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 230-235 1374894-7 1992 Similarly, haloperidol, but not clozapine, induced c-Fos-like immunoreactivity in the caudate-putamen. Haloperidol 11-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 1374894-9 1992 Like haloperidol, amphetamine induced both c-fos and zif268 mRNA in the caudate-putamen, but the anatomic patterns of induction of c-Fos-like immunoreactivity by the two drugs were dramatically different. Amphetamine 18-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 1577756-5 1992 In the present study, the phorbol ester, phorbol 12-myristate 13-acetate (PMA), stimulated c-fos transcription in a rapid and dose-dependent manner with an 800% increase in transcription following 15-30 min of addition. Phorbol Esters 26-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 1577756-5 1992 In the present study, the phorbol ester, phorbol 12-myristate 13-acetate (PMA), stimulated c-fos transcription in a rapid and dose-dependent manner with an 800% increase in transcription following 15-30 min of addition. Tetradecanoylphorbol Acetate 41-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 1577756-5 1992 In the present study, the phorbol ester, phorbol 12-myristate 13-acetate (PMA), stimulated c-fos transcription in a rapid and dose-dependent manner with an 800% increase in transcription following 15-30 min of addition. Tetradecanoylphorbol Acetate 74-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 1577756-9 1992 When H4 cells were pretreated with PMA for 24 h, there was a decrease of 20-45% in both cytosolic and membrane PKC activity and a complete loss of PMA"s induction of c-fos transcription. Tetradecanoylphorbol Acetate 35-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-171 1577756-11 1992 When cells were pretreated with PMA for 24 h, the insulin-induced increase in transcription of c-fos was reduced by 50%. Tetradecanoylphorbol Acetate 32-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 1320720-1 1992 Messenger RNA encoding the immediate early genes (IEGs) c-fos and NGFI-A was localized by in situ hybridization of specific 35S-labelled oligonucleotides to detect activated neurones in the medulla oblongata following unilateral electrical stimulation of the vagus (nX) and aortic depressor nerve (ADN), and following mechanical stimulation of the left carotid sinus (CS). Sulfur-35 124-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 1320720-1 1992 Messenger RNA encoding the immediate early genes (IEGs) c-fos and NGFI-A was localized by in situ hybridization of specific 35S-labelled oligonucleotides to detect activated neurones in the medulla oblongata following unilateral electrical stimulation of the vagus (nX) and aortic depressor nerve (ADN), and following mechanical stimulation of the left carotid sinus (CS). Oligonucleotides 137-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 1315265-4 1992 Since the paraventricular nucleus contains a large number of glucocorticoid receptor immunoreactive cells, the effect of a synthetic glucocorticoid, dexamethasone, on the induction of Fos-LI was studied. Dexamethasone 149-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 184-187 1568207-0 1992 Retinoic acid inhibition of serum-induced c-fos transcription in a fibrosarcoma cell line. Tretinoin 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 1568207-2 1992 Northern analysis revealed that both chronic (7 days) and acute (6 h) retinoic acid treatment of serum-stimulated SSV-NRK cells caused a 6-fold decrease in c-fos mRNA levels. Tretinoin 70-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 1568207-6 1992 Because it has been shown that c-fos expression plays a pivotal role in mitogenesis of quiescent fibroblasts, we conclude that the retinoic acid-mediated down-regulation of c-fos expression is a mechanism for growth inhibition in SSV-NRK cells. Tretinoin 131-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 1568207-6 1992 Because it has been shown that c-fos expression plays a pivotal role in mitogenesis of quiescent fibroblasts, we conclude that the retinoic acid-mediated down-regulation of c-fos expression is a mechanism for growth inhibition in SSV-NRK cells. Tretinoin 131-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 173-178 1315265-5 1992 Dexamethasone treatment before immobilization considerably reduced the stress-induced expression of Fos-LI in the anterior and intermediate lobe of the pituitary but did not alter the induction of Fos-LI in the paraventricular nucleus. Dexamethasone 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-103 1315265-8 1992 The reduction of stress-induced Fos-LI in the pituitary by dexamethasone is possibly due to the diminished release of CRF factor from the paraventricular neurons. Dexamethasone 59-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 1365438-4 1992 Immunocytochemical visualization of the proto-oncogene c-fos in the caudate-putamen (CPu) of lesioned rats, revealed a sparse c-fos positive nuclei in the lesioned CPu of rats treated with SKF 38393 alone, while after combined administration of MK-801 and SKF 38393 dense labelling of nuclei was found. 1,2,3,4-tetrahydroisoquinoline-7-sulfonamide 189-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-60 1569135-5 1992 The induction of c-fos immunoreactivity in cerebral cortex was also blocked by this dose of L-kynurenine. Kynurenine 92-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 1633282-0 1992 Kainic acid injection in NTS evokes hypertension and c-fos expression in spinal cord. Kainic Acid 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 1633282-1 1992 Kainic acid injected into rat nucleus tractus solitarius (NTS) caused a slowly developing hypertension, with a 2-fold increase in Fos-immunoreactive (Fos-IR) nuclei in the area of the presympathetic bulbospinal neurons in the rostral ventrolateral medulla (RVLM) and a widespread activation of sympathetic preganglionic neurons (SPN) in the spinal cord, particularly in the mid to lower thoracic cord. Kainic Acid 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-133 1633282-1 1992 Kainic acid injected into rat nucleus tractus solitarius (NTS) caused a slowly developing hypertension, with a 2-fold increase in Fos-immunoreactive (Fos-IR) nuclei in the area of the presympathetic bulbospinal neurons in the rostral ventrolateral medulla (RVLM) and a widespread activation of sympathetic preganglionic neurons (SPN) in the spinal cord, particularly in the mid to lower thoracic cord. Kainic Acid 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-153 1633282-3 1992 More than 60% of retrogradely labelled sympathoadrenal neurons in T8 were Fos-IR after kainic acid injection, consistent with the 60-fold increases in plasma adrenaline levels observed in these rats. Kainic Acid 87-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 1511271-4 1992 Both unconditioned and conditioned stressors increased c-fos mRNA levels in the locus ceruleus which correlated with stress-induced plasma corticosterone concentrations. Corticosterone 139-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 1511275-0 1992 Fos-like immunoreactivity in the rat superficial dorsal horn induced by formalin injection in the forepaw: effects of dorsal rhizotomies. Formaldehyde 72-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 1511275-1 1992 As previously described at the lumbar spinal level, we found that 2 h after subcutaneous formalin injection in the distal part of the fore-limb, Fos-like immunoreactivity (FLI) was induced in the ipsilateral cervical enlargement. Formaldehyde 89-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-148 1608534-1 1992 C-fos mRNA expression by stimulation with subcutaneous (s.c.) administration of saline or cycloheximide (CHX) was examined in the rat striatum with or without pretreatment with ceruletide, an analogue of cholecystokinin. Ceruletide 177-187 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 1608534-2 1992 The c-fos mRNA induction 1 h after CHX stimulation (25 mg/kg, s.c.) was significantly suppressed by ceruletide pretreatment (80 micrograms/kg, s.c.) 2 h before CHX stimulation in the striatum, and tended to be suppressed by ceruletide pretreatment 4 h before saline or CHX stimulation. Cycloheximide 35-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 1608534-2 1992 The c-fos mRNA induction 1 h after CHX stimulation (25 mg/kg, s.c.) was significantly suppressed by ceruletide pretreatment (80 micrograms/kg, s.c.) 2 h before CHX stimulation in the striatum, and tended to be suppressed by ceruletide pretreatment 4 h before saline or CHX stimulation. Ceruletide 100-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 1608534-2 1992 The c-fos mRNA induction 1 h after CHX stimulation (25 mg/kg, s.c.) was significantly suppressed by ceruletide pretreatment (80 micrograms/kg, s.c.) 2 h before CHX stimulation in the striatum, and tended to be suppressed by ceruletide pretreatment 4 h before saline or CHX stimulation. Cycloheximide 160-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 1608534-2 1992 The c-fos mRNA induction 1 h after CHX stimulation (25 mg/kg, s.c.) was significantly suppressed by ceruletide pretreatment (80 micrograms/kg, s.c.) 2 h before CHX stimulation in the striatum, and tended to be suppressed by ceruletide pretreatment 4 h before saline or CHX stimulation. Ceruletide 224-234 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 1608534-2 1992 The c-fos mRNA induction 1 h after CHX stimulation (25 mg/kg, s.c.) was significantly suppressed by ceruletide pretreatment (80 micrograms/kg, s.c.) 2 h before CHX stimulation in the striatum, and tended to be suppressed by ceruletide pretreatment 4 h before saline or CHX stimulation. Sodium Chloride 259-265 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 1608534-2 1992 The c-fos mRNA induction 1 h after CHX stimulation (25 mg/kg, s.c.) was significantly suppressed by ceruletide pretreatment (80 micrograms/kg, s.c.) 2 h before CHX stimulation in the striatum, and tended to be suppressed by ceruletide pretreatment 4 h before saline or CHX stimulation. Cycloheximide 160-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 1608534-4 1992 The present findings together with previous reports suggest that ceruletide might have late onset and long-lasting suppressive effects on the expression of c-fos mRNA in the striatum and that these effects might be related to its effects on DAergic neuronal transmission. Ceruletide 65-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 1317778-8 1992 Pretreatment with N-[2-(p-bromocinnamylamino)ethyl]-5-isoquinoline-sulfonamide (H-89), a selective inhibitor of cAMP-dependent protein kinase, suppressed both basal and ACTH-increased c-fos mRNA. n-[2-(p-bromocinnamylamino)ethyl]-5-isoquinoline-sulfonamide 18-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 184-189 1317778-8 1992 Pretreatment with N-[2-(p-bromocinnamylamino)ethyl]-5-isoquinoline-sulfonamide (H-89), a selective inhibitor of cAMP-dependent protein kinase, suppressed both basal and ACTH-increased c-fos mRNA. N-(2-(4-bromocinnamylamino)ethyl)-5-isoquinolinesulfonamide 80-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 184-189 1317778-8 1992 Pretreatment with N-[2-(p-bromocinnamylamino)ethyl]-5-isoquinoline-sulfonamide (H-89), a selective inhibitor of cAMP-dependent protein kinase, suppressed both basal and ACTH-increased c-fos mRNA. Cyclic AMP 112-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 184-189 1317778-11 1992 Furthermore, pretreatment with cycloheximide (5 micrograms/ml) increased both basal and ACTH-increased c-fos mRNA. Cycloheximide 31-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 1608534-0 1992 Late onset and long-lasting suppressive effects of ceruletide, an analogue of cholecystokinin, on c-fos mRNA expression in the rat striatum. Ceruletide 51-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 1608534-1 1992 C-fos mRNA expression by stimulation with subcutaneous (s.c.) administration of saline or cycloheximide (CHX) was examined in the rat striatum with or without pretreatment with ceruletide, an analogue of cholecystokinin. Sodium Chloride 80-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 1608534-1 1992 C-fos mRNA expression by stimulation with subcutaneous (s.c.) administration of saline or cycloheximide (CHX) was examined in the rat striatum with or without pretreatment with ceruletide, an analogue of cholecystokinin. Cycloheximide 90-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 1608534-1 1992 C-fos mRNA expression by stimulation with subcutaneous (s.c.) administration of saline or cycloheximide (CHX) was examined in the rat striatum with or without pretreatment with ceruletide, an analogue of cholecystokinin. Cycloheximide 105-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 1521149-7 1992 The suppression of c-fos expression induced by 4 Hz EA was completely reversed by prior treatment with naloxone. Naloxone 103-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 21554592-4 1992 To accomplish this aim, we determined the relationship between the number of LHRH neurons expressing cFos and LH concentrations during the ascending limb of the proestrous LH surge. Luteinizing Hormone 77-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-105 21554592-5 1992 During the estrous cycle in the rat, on the afternoon of proestrus, the number of LHRH neurons expressing cFos increased as plasma LH levels increased to reach peak concentrations. Luteinizing Hormone 82-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-110 21554592-7 1992 Treatment with RU486 to block progesterone"s action on the afternoon of proestrus significantly reduced both the number of LHRH neurons expressing cFos and the magnitude of LH secretion during the entire ascending phase of the LH surge. Mifepristone 15-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-151 21554592-7 1992 Treatment with RU486 to block progesterone"s action on the afternoon of proestrus significantly reduced both the number of LHRH neurons expressing cFos and the magnitude of LH secretion during the entire ascending phase of the LH surge. Luteinizing Hormone 123-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-151 21554592-9 1992 These data, together with the demonstration of an overall reduction of cFos intensity following removal of progesterone"s actions, suggest progesterone alters the dynamics of LHRH neuronal activation by significantly reducing the recruitment of LHRH neurons and suppressing the level of activation of individual LHRH neurons. Progesterone 139-151 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-75 1374838-7 1992 The effect of the DHPs was stereospecific; (+)Bay K 8644, a Ca2+ antagonist, inhibited PMA-induced increases in c-fos and NGFI-A mRNAs. 1,4-dihydropyridine 18-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 1374838-7 1992 The effect of the DHPs was stereospecific; (+)Bay K 8644, a Ca2+ antagonist, inhibited PMA-induced increases in c-fos and NGFI-A mRNAs. Tetradecanoylphorbol Acetate 87-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 1380685-0 1992 Effect of gamma-hexachlorocyclohexane and its isomers on proto-oncogene c-fos expression in brain. Hexachlorocyclohexane 10-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-77 1380685-2 1992 The organochlorine insecticide gamma-hexachlorocyclohexane (lindane) has been shown to induce c-fos expression in a dose dependent manner. Hydrocarbons, Chlorinated 4-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 1380685-2 1992 The organochlorine insecticide gamma-hexachlorocyclohexane (lindane) has been shown to induce c-fos expression in a dose dependent manner. Hexachlorocyclohexane 31-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 1380685-2 1992 The organochlorine insecticide gamma-hexachlorocyclohexane (lindane) has been shown to induce c-fos expression in a dose dependent manner. Hexachlorocyclohexane 60-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 1380685-3 1992 30 mg/kg of lindane increased c-fos expression in cortical and hippocampal areas. Hexachlorocyclohexane 12-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 1561308-7 1992 The combination of irradiation and isoproterenol had an additional effect on the levels of c-fos and jun B mRNAs and proteins particularly at earlier experimental times (1 to 8 h). Isoproterenol 35-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 1561308-8 1992 Isoproterenol alone induced high levels of c-fos and jun B mRNA but not of c-jun mRNA. Isoproterenol 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 1561308-10 1992 These observations suggest that the expression of the proto-oncogenes c-fos, c-jun, and jun B is probably regulated through differential signal transduction pathways which may be activated by these external stimuli and may be associated with functional changes induced in the rat parotid gland by ionizing radiation and by ionizing radiation and isoproterenol. Isoproterenol 346-359 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 1584455-1 1992 We here report odor-induced mapping patterns of c-fos-like protein (Fos) immunoreactivity in the rat olfactory bulb under urethane anesthesia. Urethane 122-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-66 1584455-1 1992 We here report odor-induced mapping patterns of c-fos-like protein (Fos) immunoreactivity in the rat olfactory bulb under urethane anesthesia. Urethane 122-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 1355445-1 1992 Brain structures activated during ethanol withdrawal have been mapped by visualizing c-fos mRNA expression. Ethanol 34-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 1355445-2 1992 The regional distribution of c-fos mRNA in brain during ethanol withdrawal can be mimicked by acute injection of N-methyl-D-aspartic acid (NMDA) and is stereospecifically blocked by the NMDA receptor antagonist, MK-801. Ethanol 56-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 1355445-2 1992 The regional distribution of c-fos mRNA in brain during ethanol withdrawal can be mimicked by acute injection of N-methyl-D-aspartic acid (NMDA) and is stereospecifically blocked by the NMDA receptor antagonist, MK-801. N-Methylaspartate 113-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 1355445-2 1992 The regional distribution of c-fos mRNA in brain during ethanol withdrawal can be mimicked by acute injection of N-methyl-D-aspartic acid (NMDA) and is stereospecifically blocked by the NMDA receptor antagonist, MK-801. N-Methylaspartate 139-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 1355445-2 1992 The regional distribution of c-fos mRNA in brain during ethanol withdrawal can be mimicked by acute injection of N-methyl-D-aspartic acid (NMDA) and is stereospecifically blocked by the NMDA receptor antagonist, MK-801. Dizocilpine Maleate 212-218 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 1379495-3 1992 Phenylephrine infusion in this preparation induces c-fos expression whether the heart is beating or reversibly or irreversibly arrested by solutions enriched in KCl. Phenylephrine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 1379495-3 1992 Phenylephrine infusion in this preparation induces c-fos expression whether the heart is beating or reversibly or irreversibly arrested by solutions enriched in KCl. Potassium Chloride 161-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 1315920-6 1992 Treating JS1 cells with cycloheximide (CHX), an inhibitor of protein synthesis that commonly potentiates induction of early response genes by presumably inhibiting synthesis of transcriptional repressors, markedly induces the transcription of NGFI-A and c-fos as well as p75NGFR genes. Cycloheximide 24-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 254-259 1365438-4 1992 Immunocytochemical visualization of the proto-oncogene c-fos in the caudate-putamen (CPu) of lesioned rats, revealed a sparse c-fos positive nuclei in the lesioned CPu of rats treated with SKF 38393 alone, while after combined administration of MK-801 and SKF 38393 dense labelling of nuclei was found. 1,2,3,4-tetrahydroisoquinoline-7-sulfonamide 189-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 1365438-4 1992 Immunocytochemical visualization of the proto-oncogene c-fos in the caudate-putamen (CPu) of lesioned rats, revealed a sparse c-fos positive nuclei in the lesioned CPu of rats treated with SKF 38393 alone, while after combined administration of MK-801 and SKF 38393 dense labelling of nuclei was found. Dizocilpine Maleate 245-251 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-60 1365438-4 1992 Immunocytochemical visualization of the proto-oncogene c-fos in the caudate-putamen (CPu) of lesioned rats, revealed a sparse c-fos positive nuclei in the lesioned CPu of rats treated with SKF 38393 alone, while after combined administration of MK-801 and SKF 38393 dense labelling of nuclei was found. Dizocilpine Maleate 245-251 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 1365438-4 1992 Immunocytochemical visualization of the proto-oncogene c-fos in the caudate-putamen (CPu) of lesioned rats, revealed a sparse c-fos positive nuclei in the lesioned CPu of rats treated with SKF 38393 alone, while after combined administration of MK-801 and SKF 38393 dense labelling of nuclei was found. 1,2,3,4-tetrahydroisoquinoline-7-sulfonamide 256-259 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-60 1365438-4 1992 Immunocytochemical visualization of the proto-oncogene c-fos in the caudate-putamen (CPu) of lesioned rats, revealed a sparse c-fos positive nuclei in the lesioned CPu of rats treated with SKF 38393 alone, while after combined administration of MK-801 and SKF 38393 dense labelling of nuclei was found. 1,2,3,4-tetrahydroisoquinoline-7-sulfonamide 256-259 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 1365438-5 1992 The results indicate that blockade of NMDA receptors by MK-801 acts synergistically with D-1 agonists in the induction of turning after DA denervation and shows that these changes are correlated with c-fos induction in specific areas of the CPu. Dizocilpine Maleate 56-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 200-205 1371276-2 1992 Previous experiments have shown that heparin inhibits induction of c-fos and c-myc protooncogene mRNA in rat VSMC stimulated by phorbol 12-myristate 13-acetate (PMA) but not when stimulated by epidermal growth factor (EGF) (Pukac, L. A., Castellot, J. J., Wright, T. C., Caleb, B. L., and Karnovsky, M. J. Heparin 37-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 1371276-2 1992 Previous experiments have shown that heparin inhibits induction of c-fos and c-myc protooncogene mRNA in rat VSMC stimulated by phorbol 12-myristate 13-acetate (PMA) but not when stimulated by epidermal growth factor (EGF) (Pukac, L. A., Castellot, J. J., Wright, T. C., Caleb, B. L., and Karnovsky, M. J. Tetradecanoylphorbol Acetate 128-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 1371276-6 1992 Heparin inhibited serum stimulation of c-fos mRNA in control VSMC, but heparin did not inhibit the smaller but significant serum stimulation of c-fos mRNA in PKC down-regulated VSMC, indicating that heparin may selectively inhibit PKC-dependent, but not PKC-independent, stimulation of gene expression. Heparin 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 1635664-2 1992 We have used in situ hybridization to study the pattern of c-fos expression in limbic structures following kainic acid-induced seizures during the postnatal period in the rat. Kainic Acid 107-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 1611536-3 1992 In the hypothalamus of hypertonic saline-injected rats, c-fos-mRNA positive cells were mainly restricted to the supraoptic and paraventricular nuclei and to structures associated with the lamina terminalis of the third ventricle, including in particular the subfornical organ, the organum vasculosum of the lamina terminalis (OVLT) and the median preoptic nucleus. Sodium Chloride 34-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 1322332-10 1992 Although both FSH and forskolin activated c-fos and PPenk gene expression in Sertoli cells, the germ cell factor(s) that stimulated PPenk mRNA levels did not affect the expression of this oncogene. Colforsin 22-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 1737937-3 1992 The inducible nuclear proteins binding to this site have the characteristics of AP-1, as judged by their kinetics of induction, the ability to compete and be competed efficiently by a metallothionein AP-1 site oligonucleotide, and their reaction with antibodies to Fos and Jun proteins. Oligonucleotides 210-225 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 265-268 1343505-3 1992 To elucidate some of the initial events which lead to this upregulation, we studied the acute effects of dopamine D2 agonists and antagonists on the expression of c-jun and c-fos. Dopamine 105-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 173-178 1343505-5 1992 of haloperidol (2 mg/kg) produced a rapid and transient increase in c-jun and c-fos mRNA in the rat striatum. Haloperidol 3-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 1510825-8 1992 The changes in c-fos and (or) c-myc gene expression in the uterus and nonreproductive organs could be due to sexual steroids and (or) systemic factors from uterine cells or blastocysts. Steroids 116-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 1740241-7 1992 The expression of c-fos was inhibited by superoxide dismutase but not by catalase and was super-induced by cycloheximide. Cycloheximide 107-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 1740241-9 1992 Chelation of extracellular ionized calcium by EGTA or of intracellular ionized calcium by Quin 2/AM resulted in a marked decrease of c-fos expression. Calcium 35-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 1740241-9 1992 Chelation of extracellular ionized calcium by EGTA or of intracellular ionized calcium by Quin 2/AM resulted in a marked decrease of c-fos expression. Egtazic Acid 46-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 1740241-9 1992 Chelation of extracellular ionized calcium by EGTA or of intracellular ionized calcium by Quin 2/AM resulted in a marked decrease of c-fos expression. Calcium 79-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 1740241-9 1992 Chelation of extracellular ionized calcium by EGTA or of intracellular ionized calcium by Quin 2/AM resulted in a marked decrease of c-fos expression. Quin2 90-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 1740241-10 1992 Two protein kinase C inhibitors, H-7 and staurosporine, partly diminished the expression of c-fos, whereas a third, 2-aminopurine, which has a broader spectrum of inhibiting protein kinases, almost completely abolished it. 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine 33-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 1740241-10 1992 Two protein kinase C inhibitors, H-7 and staurosporine, partly diminished the expression of c-fos, whereas a third, 2-aminopurine, which has a broader spectrum of inhibiting protein kinases, almost completely abolished it. Staurosporine 41-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 1740241-13 1992 This c-fos expression appears to be largely controlled by calcium ion movement, which could include protein kinase C activation. Calcium 58-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 5-10 1309563-5 1992 The PKC activator phorbol 12-myristate 13-acetate (PMA) stimulated c-fos expression but did not trigger cell proliferation. Tetradecanoylphorbol Acetate 51-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 1543678-6 1992 Inductions of jun and fos transcripts in the uterus by estradiol exhibit similar dose response curves (maximum responses at 4 micrograms/kg). Estradiol 55-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-25 1543678-8 1992 In vivo treatment with the phorbol ester TPA rapidly elevates uterine levels of fos, jun, and myc transcripts, indicating that expression of these protooncogenes is under non-estrogenic as well as estrogenic regulation in this target tissue. Phorbol Esters 27-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 1543678-8 1992 In vivo treatment with the phorbol ester TPA rapidly elevates uterine levels of fos, jun, and myc transcripts, indicating that expression of these protooncogenes is under non-estrogenic as well as estrogenic regulation in this target tissue. Tetradecanoylphorbol Acetate 41-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 1549354-2 1992 This C-kinase-induced expression of c-fos was prevented by in vivo competition using co-injection of oligonucleotides corresponding to the sequence of either the serum response element (SRE) or the fos AP-1 binding sequence (FAP) adjacent to SRE. Oligonucleotides 101-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 1309563-5 1992 The PKC activator phorbol 12-myristate 13-acetate (PMA) stimulated c-fos expression but did not trigger cell proliferation. Tetradecanoylphorbol Acetate 18-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 1549354-4 1992 In contrast, the induction of c-fos by serum or by casein kinase II microinjection, which is also inhibited by injection of SRE oligonucleotides, is only delayed and then markedly prolonged by injecting TRE/FAP sequence, demonstrating that the FAP site plays a prominent role in vivo in the down-regulation of the endogenous c-fos gene expression. Oligonucleotides 128-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 1542426-2 1992 Cells showing Fos-like immunoreactivity were especially dense in dorsal and external cortices of the inferior colliculus, and were nearly absent after pretreatment with the N-methyl-D-aspartate (NMDA) antagonist MK-801. N-Methylaspartate 173-193 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 1542426-2 1992 Cells showing Fos-like immunoreactivity were especially dense in dorsal and external cortices of the inferior colliculus, and were nearly absent after pretreatment with the N-methyl-D-aspartate (NMDA) antagonist MK-801. N-Methylaspartate 195-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 1542426-2 1992 Cells showing Fos-like immunoreactivity were especially dense in dorsal and external cortices of the inferior colliculus, and were nearly absent after pretreatment with the N-methyl-D-aspartate (NMDA) antagonist MK-801. Dizocilpine Maleate 212-218 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 1551231-4 1992 Gangliosides were found to completely prevent the inhibition by K-252a of NGF-induced neurite regeneration and c-fos induction, and partially also that of protein kinase N activation. Gangliosides 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 1545011-0 1992 Morphine or U-50,488 suppresses Fos protein-like immunoreactivity in the spinal cord and nucleus tractus solitarii evoked by a noxious visceral stimulus in the rat. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 1545011-0 1992 Morphine or U-50,488 suppresses Fos protein-like immunoreactivity in the spinal cord and nucleus tractus solitarii evoked by a noxious visceral stimulus in the rat. u-50 12-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 1545011-3 1992 It also compared the inhibition of pain behavior and Fos expression by a mu-selective opioid agonist, morphine, and a kappa-selective opioid agonist, U-50,488. Morphine 102-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 1338562-4 1992 This accumulation of c-fos mRNA was suppressed by protein kinase C inhibitors at the transcriptional level and was inhibited markedly by down-regulation of protein kinase C. Moreover, myocyte stretching increased inositol phosphate levels. Inositol Phosphates 213-231 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 1374838-5 1992 The phorbol ester PMA did not alter T- or L-type Ca2+ current or 45Ca2+ uptake by GH4C1 cells, but triggered large increases in both c-fos and NGFI-A mRNA. Phorbol Esters 4-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 1521461-5 1992 12-O-Tetradecanoylphorbol-13-acetate (TPA)-responsive element-like sequences have been identified in upstream regions of the GST-P gene, and oncogene products c-jun and c-fos are suggested to activate the gene. Tetradecanoylphorbol Acetate 0-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-174 1312200-3 1992 Treatment of cells with dexamethasone resulted in a 10-fold decline in c-fos transcripts and a small increase in proenkephalin mRNA. Dexamethasone 24-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 1312200-4 1992 Combined exposure to dexamethasone and isoproterenol also induced a decrease in c-fos mRNA while proenkephalin mRNA increased 8-fold. Dexamethasone 21-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 1312200-4 1992 Combined exposure to dexamethasone and isoproterenol also induced a decrease in c-fos mRNA while proenkephalin mRNA increased 8-fold. Isoproterenol 39-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 1312200-5 1992 Treatment of the C6 cells with phorbol 12-myristate 13-acetate caused a 13-fold increase in c-fos expression 0.5 h after administration and a decrease in proenkephalin mRNA. Tetradecanoylphorbol Acetate 31-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 1521461-5 1992 12-O-Tetradecanoylphorbol-13-acetate (TPA)-responsive element-like sequences have been identified in upstream regions of the GST-P gene, and oncogene products c-jun and c-fos are suggested to activate the gene. Tetradecanoylphorbol Acetate 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-174 1360906-0 1992 Intrastriatal dopamine-rich grafts induce a hyperexpression of Fos protein when challenged with amphetamine. Dopamine 14-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 12106364-0 1992 Striatal c-fos Induction by Cocaine or Apomorphine Occurs Preferentially in Output Neurons Projecting to the Substantia Nigra in the Rat. Cocaine 28-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 1360906-0 1992 Intrastriatal dopamine-rich grafts induce a hyperexpression of Fos protein when challenged with amphetamine. Amphetamine 96-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 12106364-2 1992 Ten days later, striatal c-fos was induced by systemic administration of cocaine (five normal rats; 25 mg/kg cocaine i.p. Cocaine 73-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 1360906-1 1992 The aim of the present experiment was to characterize the effect of intrastriatal grafts of embryonic dopaminergic neurones on the expression of Fos protein in the striatum when challenged with amphetamine. Amphetamine 194-205 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-148 12106364-5 1992 Cocaine induced a proportionally higher c-fos expression in striato-nigral compared to striato-pallidal neurons, whereas apomorphine activated Fos almost exclusively in striato-nigral neurons. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 1360906-4 1992 Administration of d-amphetamine induced an increase in the density of Fos-positive nuclei in the intact striatum. Dextroamphetamine 18-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 12106364-5 1992 Cocaine induced a proportionally higher c-fos expression in striato-nigral compared to striato-pallidal neurons, whereas apomorphine activated Fos almost exclusively in striato-nigral neurons. Apomorphine 121-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-146 1360906-5 1992 This stimulatory effect of amphetamine on c-fos expression was blocked by 6-hydroxydopamine hydrobromide lesions and was restored in the striata bearing transplants. Amphetamine 27-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 1360906-5 1992 This stimulatory effect of amphetamine on c-fos expression was blocked by 6-hydroxydopamine hydrobromide lesions and was restored in the striata bearing transplants. Oxidopamine 74-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 1360906-7 1992 This hyperexpression of Fos-positive nuclei was correlated with the exaggerated compensation of amphetamine-induced rotation in the same animals. Amphetamine 96-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 1347413-0 1992 Dopaminergic transplants normalize amphetamine- and apomorphine-induced Fos expression in the 6-hydroxydopamine-lesioned striatum. Apomorphine 52-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 1543589-2 1992 The present experiments show that vagal sensory fibres mediate CCK-induced c-fos in brain stem neurons, based on the findings that perivagal capsaicin pre-treatment in seven out of eight cases reduced or ablated c-fos expression in these regions. Capsaicin 141-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 1543589-2 1992 The present experiments show that vagal sensory fibres mediate CCK-induced c-fos in brain stem neurons, based on the findings that perivagal capsaicin pre-treatment in seven out of eight cases reduced or ablated c-fos expression in these regions. Capsaicin 141-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 212-217 1285948-0 1992 Opposite effects of rough and gentle handling with repeated saline administration on c-fos mRNA expression in the rat brain. Sodium Chloride 60-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 1285948-1 1992 The rough handling with repeated saline administration (1.2 ml/kg s.c. for 7 days) enhanced cortical c-fos mRNA expression in the rat brain after a single saline stimulation (1.2 ml/kg s.c.) due to increasing baseline c-fos mRNA levels, whereas the gentle handling with repeated saline administration declined c-fos mRNA expression after a single injection due to decreasing the baseline of c-fos mRNA levels. Sodium Chloride 33-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 1285948-1 1992 The rough handling with repeated saline administration (1.2 ml/kg s.c. for 7 days) enhanced cortical c-fos mRNA expression in the rat brain after a single saline stimulation (1.2 ml/kg s.c.) due to increasing baseline c-fos mRNA levels, whereas the gentle handling with repeated saline administration declined c-fos mRNA expression after a single injection due to decreasing the baseline of c-fos mRNA levels. Sodium Chloride 33-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 218-223 1285948-1 1992 The rough handling with repeated saline administration (1.2 ml/kg s.c. for 7 days) enhanced cortical c-fos mRNA expression in the rat brain after a single saline stimulation (1.2 ml/kg s.c.) due to increasing baseline c-fos mRNA levels, whereas the gentle handling with repeated saline administration declined c-fos mRNA expression after a single injection due to decreasing the baseline of c-fos mRNA levels. Sodium Chloride 33-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 218-223 1285948-1 1992 The rough handling with repeated saline administration (1.2 ml/kg s.c. for 7 days) enhanced cortical c-fos mRNA expression in the rat brain after a single saline stimulation (1.2 ml/kg s.c.) due to increasing baseline c-fos mRNA levels, whereas the gentle handling with repeated saline administration declined c-fos mRNA expression after a single injection due to decreasing the baseline of c-fos mRNA levels. Sodium Chloride 33-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 218-223 1285948-1 1992 The rough handling with repeated saline administration (1.2 ml/kg s.c. for 7 days) enhanced cortical c-fos mRNA expression in the rat brain after a single saline stimulation (1.2 ml/kg s.c.) due to increasing baseline c-fos mRNA levels, whereas the gentle handling with repeated saline administration declined c-fos mRNA expression after a single injection due to decreasing the baseline of c-fos mRNA levels. Sodium Chloride 155-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 1285948-1 1992 The rough handling with repeated saline administration (1.2 ml/kg s.c. for 7 days) enhanced cortical c-fos mRNA expression in the rat brain after a single saline stimulation (1.2 ml/kg s.c.) due to increasing baseline c-fos mRNA levels, whereas the gentle handling with repeated saline administration declined c-fos mRNA expression after a single injection due to decreasing the baseline of c-fos mRNA levels. Sodium Chloride 155-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 1285949-0 1992 Muscarinic cholinergic receptor-mediated modulation on striatal c-fos mRNA expression induced by levodopa in rat brain. Levodopa 97-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 1285949-3 1992 Levodopa given alone increase the expression of c-fos mRNA. Levodopa 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 1285949-4 1992 Although carbachol or trihexyphenidyl alone was ineffective in inducing c-fos mRNA, the combination of levodopa and carbachol (> or = to 0.1 mg/kg) significantly suppressed the induction of c-fos mRNA as compared with levodopa given alone. Levodopa 103-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-198 1285949-4 1992 Although carbachol or trihexyphenidyl alone was ineffective in inducing c-fos mRNA, the combination of levodopa and carbachol (> or = to 0.1 mg/kg) significantly suppressed the induction of c-fos mRNA as compared with levodopa given alone. Carbachol 116-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-198 1285949-4 1992 Although carbachol or trihexyphenidyl alone was ineffective in inducing c-fos mRNA, the combination of levodopa and carbachol (> or = to 0.1 mg/kg) significantly suppressed the induction of c-fos mRNA as compared with levodopa given alone. Levodopa 221-229 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-198 1285949-5 1992 The combined administration of levodopa and trihexyphenidyl showed a trend toward an additive effect on the induction of c-fos mRNA vs levodopa alone. Levodopa 31-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-126 1285949-5 1992 The combined administration of levodopa and trihexyphenidyl showed a trend toward an additive effect on the induction of c-fos mRNA vs levodopa alone. Trihexyphenidyl 44-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-126 1285949-5 1992 The combined administration of levodopa and trihexyphenidyl showed a trend toward an additive effect on the induction of c-fos mRNA vs levodopa alone. Levodopa 135-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-126 1285949-6 1992 These findings suggest that the muscarinic cholinergic system may modulate the levodopa-induced c-fos mRNA expression which then regulates the expression of other mRNAs. Levodopa 79-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 1347413-0 1992 Dopaminergic transplants normalize amphetamine- and apomorphine-induced Fos expression in the 6-hydroxydopamine-lesioned striatum. Oxidopamine 94-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 1347413-2 1992 The pattern of striatal Fos expression after systemic administration of either the dopamine receptor agonist, apomorphine, or the dopamine-releasing agent, amphetamine, was studied in rats which had received cell suspension grafts of fetal ventral mesencephalic neurons into the striatum after a complete 6-hydroxydopamine lesion of mesostriatal dopaminergic projection. Apomorphine 110-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 1347413-2 1992 The pattern of striatal Fos expression after systemic administration of either the dopamine receptor agonist, apomorphine, or the dopamine-releasing agent, amphetamine, was studied in rats which had received cell suspension grafts of fetal ventral mesencephalic neurons into the striatum after a complete 6-hydroxydopamine lesion of mesostriatal dopaminergic projection. Amphetamine 156-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 1347413-6 1992 Consistent with previous studies, amphetamine induced high Fos expression in the medial and dorsal parts of the intact caudate-putamen and significantly lower expression in the denervated caudate-putamen. Amphetamine 34-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 1347413-8 1992 In grafted rats, amphetamine-induced Fos activation was restored to normal or supranormal levels in the anterior and central caudate-putamen (i.e. close to the graft deposits), whereas in the tail of caudate-putamen Fos expression was significantly lower than normal. Amphetamine 17-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 1347413-8 1992 In grafted rats, amphetamine-induced Fos activation was restored to normal or supranormal levels in the anterior and central caudate-putamen (i.e. close to the graft deposits), whereas in the tail of caudate-putamen Fos expression was significantly lower than normal. Amphetamine 17-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 216-219 1347413-10 1992 Apomorphine led to high Fos activation throughout the dopamine-depleted caudate-putamen, whereas only very few immunopositive cells were observed in the intact caudate-putamen. Apomorphine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 1347413-12 1992 In the grafted rats, apomorphine-induced Fos activation was similar to normal in all striatal areas sampled, as well as in the globus pallidus. Apomorphine 21-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 1815818-1 1991 The protooncogene c-fos is expressed rapidly, transiently and polysynaptically within neurons in response to synaptic activation and voltage-gated calcium entry into the cell. Calcium 147-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 1347407-5 1992 That many of the Fos-positive neurons project to the substantia nigra was confirmed by retrograde labeling with Fluoro-Gold. fluoro- 112-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 1347413-0 1992 Dopaminergic transplants normalize amphetamine- and apomorphine-induced Fos expression in the 6-hydroxydopamine-lesioned striatum. Amphetamine 35-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 1954900-11 1991 The time course for the effect of thyroidectomy on Fos expression in the pPVN paralleled increased plasma TSH concentration. Thyrotropin 106-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 1668121-0 1991 Platelet-activating factor and polyunsaturated fatty acids in cerebral ischemia or convulsions: intracellular PAF-binding sites and activation of a fos/jun/AP-1 transcriptional signaling system. Fatty Acids, Unsaturated 31-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-151 1686645-7 1991 In contrast, c-fos mRNA was induced in most hippocampal and cortical neurons starting 1.5 h after the kainic acid injection. Kainic Acid 102-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 1686849-0 1991 Differential effects of reserpine on brainstem catecholaminergic neurons revealed by Fos protein immunohistochemistry. Reserpine 24-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 1773343-2 1991 Immunohistochemical analysis, using a polyclonal antibody for Fos, revealed a distinct pattern of neuronal activation in the off-axis animals in the dorsomedial cell column (DMCC) of the inferior olivary nucleus. dmcc 174-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 1686849-1 1991 The effects of a single systemic injection of reserpine on c-fos proto-oncogene expression in catecholaminergic neurons of the rat brainstem were studied by immunohistochemistry for Fos proteins (Fos). Reserpine 46-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 1686849-1 1991 The effects of a single systemic injection of reserpine on c-fos proto-oncogene expression in catecholaminergic neurons of the rat brainstem were studied by immunohistochemistry for Fos proteins (Fos). Reserpine 46-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 182-185 1686849-1 1991 The effects of a single systemic injection of reserpine on c-fos proto-oncogene expression in catecholaminergic neurons of the rat brainstem were studied by immunohistochemistry for Fos proteins (Fos). Reserpine 46-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 196-199 1686849-11 1991 The results demonstrate that acute reserpine treatment induces Fos expression in distinct populations of brainstem neurons, comprising both catecholaminergic and non-catecholaminergic neurons. Reserpine 35-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 1686849-12 1991 Thus, induction of Fos by reserpine does not coincide with the site of action of this drug. Reserpine 26-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 1686849-13 1991 The distribution of Fos immunoreactive NA neurons after reserpine treatment is comparable to that reported after application of stressful stimuli. Reserpine 56-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 1681906-1 1991 The beta-adrenoreceptor agonist isoproterenol elevates cAMP concentrations in the A5 rat salivary epithelial cell line and rapidly and transiently induces the expression of c-fos and jun B at 30 and 60 min following continuous stimulation of these cells. Isoproterenol 32-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 173-178 1681906-3 1991 We show here that the inducibility of these genes by isoproterenol or 8-BrcAMP is transcriptionally regulated and short (5 min) incubations of A5 cells with either agent is sufficient to trigger the induction of c-fos and jun B. Isoproterenol 53-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 212-217 1681906-7 1991 These studies provide the first evidence for the transcriptional regulation of c-fos and jun B by beta-adrenergic receptor stimulation or cAMP in an epithelial cell line (A5) and demonstrate the coordinate expression and inducibility of these genes at the different stages of the A5 cell cycle. Cyclic AMP 138-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 1797341-0 1991 Intravenous hypertonic saline induces Fos immunoreactivity in neurons throughout the lamina terminalis. Sodium Chloride 23-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 1797341-2 1991 Intravenous 1.5 or 0.75 mol/l NaCl or 1.2 mol/l sucrose in 0.15 mol/l NaCl, but not isotonic 0.15 mol/l NaCl, caused increased Fos expression in the hypothalamic paraventricular and supraoptic nuclei and throughout the lamina terminalis (organum vasculosum laminae terminalis, median preoptic nucleus and subfornical organ). Sodium Chloride 30-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-130 1797341-2 1991 Intravenous 1.5 or 0.75 mol/l NaCl or 1.2 mol/l sucrose in 0.15 mol/l NaCl, but not isotonic 0.15 mol/l NaCl, caused increased Fos expression in the hypothalamic paraventricular and supraoptic nuclei and throughout the lamina terminalis (organum vasculosum laminae terminalis, median preoptic nucleus and subfornical organ). Sucrose 48-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-130 1797341-2 1991 Intravenous 1.5 or 0.75 mol/l NaCl or 1.2 mol/l sucrose in 0.15 mol/l NaCl, but not isotonic 0.15 mol/l NaCl, caused increased Fos expression in the hypothalamic paraventricular and supraoptic nuclei and throughout the lamina terminalis (organum vasculosum laminae terminalis, median preoptic nucleus and subfornical organ). Sodium Chloride 70-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-130 1797341-2 1991 Intravenous 1.5 or 0.75 mol/l NaCl or 1.2 mol/l sucrose in 0.15 mol/l NaCl, but not isotonic 0.15 mol/l NaCl, caused increased Fos expression in the hypothalamic paraventricular and supraoptic nuclei and throughout the lamina terminalis (organum vasculosum laminae terminalis, median preoptic nucleus and subfornical organ). Sodium Chloride 70-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-130 1772009-0 1991 Post-ischemic and kainic acid-induced c-fos protein expression in the rat hippocampus. Kainic Acid 18-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 1915270-2 1991 The induction of c-fos by CKII is inhibited by coinjection of oligonucleotides corresponding to the sequence of the serum response element (SRE) present in the c-fos promoter, indicating that competitive displacement of positive factors from the endogenous c-fos SRE prevents c-fos induction by CKII. Oligonucleotides 62-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 1915270-2 1991 The induction of c-fos by CKII is inhibited by coinjection of oligonucleotides corresponding to the sequence of the serum response element (SRE) present in the c-fos promoter, indicating that competitive displacement of positive factors from the endogenous c-fos SRE prevents c-fos induction by CKII. Oligonucleotides 62-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 1915270-2 1991 The induction of c-fos by CKII is inhibited by coinjection of oligonucleotides corresponding to the sequence of the serum response element (SRE) present in the c-fos promoter, indicating that competitive displacement of positive factors from the endogenous c-fos SRE prevents c-fos induction by CKII. Oligonucleotides 62-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 1915270-2 1991 The induction of c-fos by CKII is inhibited by coinjection of oligonucleotides corresponding to the sequence of the serum response element (SRE) present in the c-fos promoter, indicating that competitive displacement of positive factors from the endogenous c-fos SRE prevents c-fos induction by CKII. Oligonucleotides 62-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 1915733-4 1991 The rapid increase in BDNF and NGF mRNA after a needle insertion or injection of saline was transient and preceded by an increase in c-fos mRNA in the same brain regions. Sodium Chloride 81-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 1726120-0 1991 Enkephalin, substance P, and serotonin axonal input to c-fos-like immunoreactive neurons of the rat spinal cord. Serotonin 29-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 1726120-1 1991 Mustard oil, which stimulates small diameter afferents, was used to evoke the expression of the oncogene c-fos in the lumbar spinal cord. mustard oil 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 1726120-4 1991 The analysis of vibratome and semithin plastic-embedded tissue sections demonstrated that the majority of c-fos-like immunoreactive neurons received input from enkephalin-, substance P- or serotonin-immunoreactive axonal varicosities. Serotonin 189-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 1655570-0 1991 cAMP stimulates the C/EBP-related transcription factor rNFIL-6 to trans-locate to the nucleus and induce c-fos transcription. Cyclic AMP 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 1655570-6 1991 These data show that rNFIL-6 undergoes a novel activation in which cAMP-induced nuclear trans-location allows rNFIL-6 to bind to the SRE and contribute to c-fos activation. Cyclic AMP 67-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-160 1655570-7 1991 We propose that rNFIL-6 is an additional regulatory component of the c-fos gene, which provides cAMP responsiveness to the multifunctional SRE. Cyclic AMP 96-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 1805451-1 1991 We have previously shown that the intracellular content of c-fos mRNA is rapidly induced (within 1 to 3 hours) in ovariectomized rat or mouse uteri following administration of estradiol. Estradiol 176-185 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 1910045-2 1991 Recently, it has been demonstrated that the same cis-acting element in the 5" region of the c-fos gene has the ability to mediate both cAMP- and calcium-induced c-fos expression in PC12 cells (Sheng, M., McFadden, G., and Greenberg, M. (1990) Neuron 4, 571-582). Cyclic AMP 135-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 1910045-2 1991 Recently, it has been demonstrated that the same cis-acting element in the 5" region of the c-fos gene has the ability to mediate both cAMP- and calcium-induced c-fos expression in PC12 cells (Sheng, M., McFadden, G., and Greenberg, M. (1990) Neuron 4, 571-582). Cyclic AMP 135-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 1910045-2 1991 Recently, it has been demonstrated that the same cis-acting element in the 5" region of the c-fos gene has the ability to mediate both cAMP- and calcium-induced c-fos expression in PC12 cells (Sheng, M., McFadden, G., and Greenberg, M. (1990) Neuron 4, 571-582). Calcium 145-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 1910045-2 1991 Recently, it has been demonstrated that the same cis-acting element in the 5" region of the c-fos gene has the ability to mediate both cAMP- and calcium-induced c-fos expression in PC12 cells (Sheng, M., McFadden, G., and Greenberg, M. (1990) Neuron 4, 571-582). Calcium 145-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 1910045-3 1991 Here we demonstrate that both cAMP- and calcium-mediated induction of c-fos and egr1 are dependent on PKA activity. Cyclic AMP 30-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 1910045-3 1991 Here we demonstrate that both cAMP- and calcium-mediated induction of c-fos and egr1 are dependent on PKA activity. Calcium 40-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 1910045-4 1991 Addition of either depolarizing concentrations of KCl or the calcium ionophore, ionomycin, to PC12 cells increased the expression of both c-fos and egr1, but these inductions were dramatically reduced in three PKA-deficient cell lines, 123.7, AB.11, and A126-1B2. Potassium Chloride 50-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 1910045-4 1991 Addition of either depolarizing concentrations of KCl or the calcium ionophore, ionomycin, to PC12 cells increased the expression of both c-fos and egr1, but these inductions were dramatically reduced in three PKA-deficient cell lines, 123.7, AB.11, and A126-1B2. Calcium 61-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 1910045-4 1991 Addition of either depolarizing concentrations of KCl or the calcium ionophore, ionomycin, to PC12 cells increased the expression of both c-fos and egr1, but these inductions were dramatically reduced in three PKA-deficient cell lines, 123.7, AB.11, and A126-1B2. Ionomycin 80-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 1910045-5 1991 Furthermore, pretreatment of PC12 cells with 20 microM H89, a specific inhibitor of PKA, inhibited forskolin, dibutyryl cAMP, and KCl-induced c-fos and egr1 induction, while having no effect on NGF induction. Colforsin 99-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-147 1910045-5 1991 Furthermore, pretreatment of PC12 cells with 20 microM H89, a specific inhibitor of PKA, inhibited forskolin, dibutyryl cAMP, and KCl-induced c-fos and egr1 induction, while having no effect on NGF induction. Potassium Chloride 130-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-147 1745400-1 1991 Electrical stimulation (20-35 Hz, 2-5 V, 1.5 h) of the pelvic nerve in urethane-anesthetized rats increased the expression of c-fos protein-immunoreactivity primarily in neurons in the L6-S1 segments of the spinal cord. Urethane 71-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 1908349-4 1991 The data demonstrate that BBI is capable of preventing radiation-induced overexpression of c-myc and c-fos without interfering with the constitutive expression of these 2 genes. Amiodarone 26-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 1908349-6 1991 The data demonstrate that the anticarcinogenic BBI selectively inhibits the overexpression of c-myc and c-fos while not affecting crypt cell proliferation. Amiodarone 47-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 1908349-7 1991 These results suggest that a protease is involved in the pathway for enhanced c-myc and c-fos expression and that protease inhibitors such as BBI can interrupt this pathway. Amiodarone 142-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 1683539-1 1991 Focus on functional compartments produced by d-amphetamine activation of the c-fos gene and its relationship to the glucocorticoid receptor. Dextroamphetamine 45-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 1683539-2 1991 A new computer-assisted image analysis procedure was developed for the analysis of striatal compartments of the rats visualized in this case by means of c-fos immunocytochemistry after morphine and/or d-amphetamine treatments. Morphine 185-193 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-158 1683539-3 1991 In particular it has been shown that d-amphetamine can induce an activation of the c-fos early gene in various subregions of the neostriatum and that morphine treatment can antagonize this activation. Dextroamphetamine 37-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 1683539-3 1991 In particular it has been shown that d-amphetamine can induce an activation of the c-fos early gene in various subregions of the neostriatum and that morphine treatment can antagonize this activation. Morphine 150-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 1890812-1 1991 Sequential and transient expression of c-fos, c-jun, c-myc, c-Ha-ras and c-Ki-ras proto-oncogene RNA transcripts with zonal heterogeneity was demonstrated in virtually all hepatocytes of adult rat liver by in situ hybridization with single-stranded, [35S]-labeled cRNA probes at various time points after intraperitoneal administration of a single dose of carbon tetrachloride (CCl4). Sulfur-35 251-254 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 1890812-1 1991 Sequential and transient expression of c-fos, c-jun, c-myc, c-Ha-ras and c-Ki-ras proto-oncogene RNA transcripts with zonal heterogeneity was demonstrated in virtually all hepatocytes of adult rat liver by in situ hybridization with single-stranded, [35S]-labeled cRNA probes at various time points after intraperitoneal administration of a single dose of carbon tetrachloride (CCl4). Carbon Tetrachloride 356-376 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 1890812-1 1991 Sequential and transient expression of c-fos, c-jun, c-myc, c-Ha-ras and c-Ki-ras proto-oncogene RNA transcripts with zonal heterogeneity was demonstrated in virtually all hepatocytes of adult rat liver by in situ hybridization with single-stranded, [35S]-labeled cRNA probes at various time points after intraperitoneal administration of a single dose of carbon tetrachloride (CCl4). Carbon Tetrachloride 378-382 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 1659543-8 1991 However, tetradecanoyl-13-phorbol acetate (TPA, 200 nM) was able to induce c-fos mRNA expression. tetradecanoyl-13-phorbol acetate 9-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 1659543-8 1991 However, tetradecanoyl-13-phorbol acetate (TPA, 200 nM) was able to induce c-fos mRNA expression. Tetradecanoylphorbol Acetate 43-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 1659543-9 1991 The protein kinase C (PKC) inhibitors H-7 (0.3-30 microM) and staurosporine (0.75 micrograms/ml) blocked FSH-induced c-fos mRNA expression in cultured granulosa cells while HA 1004, an inhibitor of cGMP- and cAMP-dependent protein kinases at 30 microM had no effect on TPA-induced c-fos expression, and only minimally inhibited FSH-induced c-fos expression. 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 1659543-9 1991 The protein kinase C (PKC) inhibitors H-7 (0.3-30 microM) and staurosporine (0.75 micrograms/ml) blocked FSH-induced c-fos mRNA expression in cultured granulosa cells while HA 1004, an inhibitor of cGMP- and cAMP-dependent protein kinases at 30 microM had no effect on TPA-induced c-fos expression, and only minimally inhibited FSH-induced c-fos expression. 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 281-286 1659543-9 1991 The protein kinase C (PKC) inhibitors H-7 (0.3-30 microM) and staurosporine (0.75 micrograms/ml) blocked FSH-induced c-fos mRNA expression in cultured granulosa cells while HA 1004, an inhibitor of cGMP- and cAMP-dependent protein kinases at 30 microM had no effect on TPA-induced c-fos expression, and only minimally inhibited FSH-induced c-fos expression. 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 281-286 1659543-9 1991 The protein kinase C (PKC) inhibitors H-7 (0.3-30 microM) and staurosporine (0.75 micrograms/ml) blocked FSH-induced c-fos mRNA expression in cultured granulosa cells while HA 1004, an inhibitor of cGMP- and cAMP-dependent protein kinases at 30 microM had no effect on TPA-induced c-fos expression, and only minimally inhibited FSH-induced c-fos expression. Staurosporine 62-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 1659543-9 1991 The protein kinase C (PKC) inhibitors H-7 (0.3-30 microM) and staurosporine (0.75 micrograms/ml) blocked FSH-induced c-fos mRNA expression in cultured granulosa cells while HA 1004, an inhibitor of cGMP- and cAMP-dependent protein kinases at 30 microM had no effect on TPA-induced c-fos expression, and only minimally inhibited FSH-induced c-fos expression. Staurosporine 62-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 281-286 1659543-9 1991 The protein kinase C (PKC) inhibitors H-7 (0.3-30 microM) and staurosporine (0.75 micrograms/ml) blocked FSH-induced c-fos mRNA expression in cultured granulosa cells while HA 1004, an inhibitor of cGMP- and cAMP-dependent protein kinases at 30 microM had no effect on TPA-induced c-fos expression, and only minimally inhibited FSH-induced c-fos expression. Staurosporine 62-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 281-286 1659543-9 1991 The protein kinase C (PKC) inhibitors H-7 (0.3-30 microM) and staurosporine (0.75 micrograms/ml) blocked FSH-induced c-fos mRNA expression in cultured granulosa cells while HA 1004, an inhibitor of cGMP- and cAMP-dependent protein kinases at 30 microM had no effect on TPA-induced c-fos expression, and only minimally inhibited FSH-induced c-fos expression. Cyclic GMP 198-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 1659543-9 1991 The protein kinase C (PKC) inhibitors H-7 (0.3-30 microM) and staurosporine (0.75 micrograms/ml) blocked FSH-induced c-fos mRNA expression in cultured granulosa cells while HA 1004, an inhibitor of cGMP- and cAMP-dependent protein kinases at 30 microM had no effect on TPA-induced c-fos expression, and only minimally inhibited FSH-induced c-fos expression. Tetradecanoylphorbol Acetate 269-272 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 1662744-4 1991 Both yohimbine injection and stressful stimulation, two treatments causing brain norepinephrine (NE) release, were found to cause a parallel, transient activation of at least 5 IEGs (c-fos, nur77, tis-7, zif-268 and tis-21) in the rat cortex. Yohimbine 5-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 183-188 1782669-7 1991 Induction of c-fos and c-myc occurred at both temperatures after the stimulation with FBS, TPA or A23187. Tetradecanoylphorbol Acetate 91-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 1782669-7 1991 Induction of c-fos and c-myc occurred at both temperatures after the stimulation with FBS, TPA or A23187. Calcimycin 98-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 1908006-1 1991 The induction of c-fos mRNA was assessed using Northern blots and in situ hybridization in adult rats administered hypertonic saline (HS) and isotonic saline (IS). Sodium Chloride 126-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 1908006-11 1991 Isotonic saline injections induced c-fos mRNA in the PVNp, anterior hypothalamus, suprachiasmatic nucleus, cingulate gyrus, neocortex, ventral lateral septal nucleus, piriform cortex, hippocampal pyramidal and dentate granule neurons, paraventricular and intralaminar thalamic nuclei, bed nuclei of stria terminalis, cortical and medial amygdaloid nuclei, and other structures. Sodium Chloride 9-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 19912814-6 1991 Colchicine (10 muM) increased m2-mAChR mRNA level in cerebellar granule cells by 14, 33, and 63% after treatment for 2, 4, and 8 h, respectively, but markedly decreased it at 24 h. The colchicine-induced up-regulation of m2-mAChR mRNA was temporally associated with a decrease in intracellular cyclic AMP levels but an increase in c-Fos mRNA. Colchicine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 331-336 19912814-6 1991 Colchicine (10 muM) increased m2-mAChR mRNA level in cerebellar granule cells by 14, 33, and 63% after treatment for 2, 4, and 8 h, respectively, but markedly decreased it at 24 h. The colchicine-induced up-regulation of m2-mAChR mRNA was temporally associated with a decrease in intracellular cyclic AMP levels but an increase in c-Fos mRNA. Colchicine 185-195 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 331-336 19912814-9 1991 c-Fos mRNA was increased in parallel by 8 h of treatment with colchicine in a concentration-dependent manner. Colchicine 62-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 1912482-0 1991 Fos expression in GHB-induced generalized absence epilepsy in the thalamus of the rat. Sodium Oxybate 18-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 1912482-1 1991 Using the model of gamma-hydroxybutyrate (GHB)-induced generalized absence epilepsy, the present work investigated the distribution of fos oncoprotein expression in the rat thalamus with fos antibody immunohistochemistry. Sodium Oxybate 19-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-138 1912482-1 1991 Using the model of gamma-hydroxybutyrate (GHB)-induced generalized absence epilepsy, the present work investigated the distribution of fos oncoprotein expression in the rat thalamus with fos antibody immunohistochemistry. Sodium Oxybate 42-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-138 1713356-3 1991 We hypothesized that nitrone free-radical spin traps promote restoration of cytosolic ATP during reperfusion and prevent c-fos induction. nitrones 21-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-126 1713356-8 1991 A fourfold elevation of c-fos occurred in the saline-treated group (p less than 0.001). Sodium Chloride 46-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 1713356-10 1991 DMPO resulted in significantly less induction of c-fos than did NS. 5,5-dimethyl-1-pyrroline-1-oxide 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 1718537-0 1991 Lithium augments pilocarpine-induced fos gene expression in rat brain. Lithium 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 1718537-0 1991 Lithium augments pilocarpine-induced fos gene expression in rat brain. Pilocarpine 17-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 1718537-2 1991 Previous studies in PC12 pheochromocytoma cells indicate that lithium has a dramatic augmenting effect on expression of the fos proto-oncogene, a component of the AP-1 transcription factor. Lithium 62-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-127 1718537-4 1991 In particular, fos induction mediated by the m1 muscarinic receptor linked to PKC activation was found to be exquisitely sensitive to lithium enhancement. Lithium 134-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-18 1718537-6 1991 Following treatment with the muscarinic agonist pilocarpine, fos mRNA accumulates in the cortex, an effect that is blocked by the m1 antagonist pirenzepine. Pilocarpine 48-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 1718537-6 1991 Following treatment with the muscarinic agonist pilocarpine, fos mRNA accumulates in the cortex, an effect that is blocked by the m1 antagonist pirenzepine. Pirenzepine 144-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 1718537-7 1991 Rats treated with a single intraperitoneal injection of lithium chloride exhibited a substantial increase in pilocarpine-mediated fos expression. Lithium Chloride 56-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-133 1718537-7 1991 Rats treated with a single intraperitoneal injection of lithium chloride exhibited a substantial increase in pilocarpine-mediated fos expression. Pilocarpine 109-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-133 1718537-9 1991 The finding that short-term treatment with lithium enhances fos expression in the brain suggests a mechanism for its therapeutic action. Lithium 43-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 1922947-1 1991 The expression of c-fos proteins (Fos) in principal sympathetic neurons and adrenal chromaffin cells was studied immunocytochemically after a single s.c. injection of nicotine (2mg/kg). Nicotine 167-175 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 2070497-4 1991 The levels of mRNAs for the c-myc and c-fos protooncogenes in livers of rats given either MDD or the liver carcinogen, 2-acetylaminofluorene (AAF), were compared by Northern blot analysis with those in livers of animals given control diets. 2-Acetylaminofluorene 119-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 1681407-0 1991 Prolonged and selective induction of Fos-related antigen(s) in striatal neurons after 6-hydroxydopamine lesions of the rat substantia nigra pars compacta. Oxidopamine 86-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 1681407-1 1991 Unilateral 6-hydroxydopamine (6-OHDA) lesions of the rat substantia nigra lead to a large widespread and long-lasting (greater than 3 months) increased expression of Fos-related antigen(s) (FRAs) in striatal neurons ipsilateral to the side of the lesion. Oxidopamine 11-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-169 1681407-1 1991 Unilateral 6-hydroxydopamine (6-OHDA) lesions of the rat substantia nigra lead to a large widespread and long-lasting (greater than 3 months) increased expression of Fos-related antigen(s) (FRAs) in striatal neurons ipsilateral to the side of the lesion. Oxidopamine 30-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-169 1681407-2 1991 However, Fos and Jun expression were only very slightly increased in a few scattered neurons in the dopamine-denervated striatum. Dopamine 100-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 1668366-0 1991 Stress selectively increases fos protein in dopamine neurons innervating the prefrontal cortex. Dopamine 44-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 1668366-1 1991 Stress-induced alterations in expression of c-fos protein (Fos) in mesencephalic dopamine (DA) neurons of the rat were examined in order to discern which midbrain DA neurons are metabolically activated by stress. Dopamine 81-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-57 1668366-1 1991 Stress-induced alterations in expression of c-fos protein (Fos) in mesencephalic dopamine (DA) neurons of the rat were examined in order to discern which midbrain DA neurons are metabolically activated by stress. Dopamine 81-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 1668366-1 1991 Stress-induced alterations in expression of c-fos protein (Fos) in mesencephalic dopamine (DA) neurons of the rat were examined in order to discern which midbrain DA neurons are metabolically activated by stress. Dopamine 91-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-57 1668366-1 1991 Stress-induced alterations in expression of c-fos protein (Fos) in mesencephalic dopamine (DA) neurons of the rat were examined in order to discern which midbrain DA neurons are metabolically activated by stress. Dopamine 91-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 1668366-4 1991 Administration of the anxiogenic beta-carboline FG 7142 also increased the total number of VTA DA neurons expressing Fos protein, whereas pretreatment with an anxiolytic benzodiazepine (diazepam) partially prevented the stress-induced increase in Fos expression. norharman 33-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 1668366-4 1991 Administration of the anxiogenic beta-carboline FG 7142 also increased the total number of VTA DA neurons expressing Fos protein, whereas pretreatment with an anxiolytic benzodiazepine (diazepam) partially prevented the stress-induced increase in Fos expression. norharman 33-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 247-250 1668366-4 1991 Administration of the anxiogenic beta-carboline FG 7142 also increased the total number of VTA DA neurons expressing Fos protein, whereas pretreatment with an anxiolytic benzodiazepine (diazepam) partially prevented the stress-induced increase in Fos expression. Benzodiazepines 170-184 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 247-250 1668366-4 1991 Administration of the anxiogenic beta-carboline FG 7142 also increased the total number of VTA DA neurons expressing Fos protein, whereas pretreatment with an anxiolytic benzodiazepine (diazepam) partially prevented the stress-induced increase in Fos expression. Diazepam 186-194 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 247-250 1831151-0 1991 Striatal c-fos induction by drugs and stress in neonatally dopamine-depleted rats given nigral transplants: importance of NMDA activation and relevance to sensitization phenomena. Dopamine 59-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 1831151-1 1991 Induction of the proto-oncogene c-fos occurred in cells of the striatum in response to stimuli that are known to release dopamine (DA). Dopamine 121-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-37 1831151-1 1991 Induction of the proto-oncogene c-fos occurred in cells of the striatum in response to stimuli that are known to release dopamine (DA). Dopamine 131-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-37 1831151-2 1991 As revealed by fos immunocytochemistry, amphetamine (AMPH) produced c-fos induction in many cells of the medial two-thirds of the striatum of normal rats, with patchy labeling in the lateral third. Amphetamine 40-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 1831151-2 1991 As revealed by fos immunocytochemistry, amphetamine (AMPH) produced c-fos induction in many cells of the medial two-thirds of the striatum of normal rats, with patchy labeling in the lateral third. Amphetamine 53-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 1831151-6 1991 The DA D1 antagonist SCH 23390 completely blocked c-fos induction in all animals. SCH 23390 21-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 1831151-7 1991 The N-methyl-D-aspartate antagonist MK-801 also blocked c-fos induction by AMPH within the medial striatum, but intensified c-fos induction laterally in those animals with DA innervation. N-Methylaspartate 4-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 1831151-7 1991 The N-methyl-D-aspartate antagonist MK-801 also blocked c-fos induction by AMPH within the medial striatum, but intensified c-fos induction laterally in those animals with DA innervation. Dizocilpine Maleate 36-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 1831151-7 1991 The N-methyl-D-aspartate antagonist MK-801 also blocked c-fos induction by AMPH within the medial striatum, but intensified c-fos induction laterally in those animals with DA innervation. Dizocilpine Maleate 36-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 1831151-9 1991 Both AMPH and stress produced turning, but only AMPH produced widespread c-fos induction, and stress-induced turning only occurred after exposure to AMPH. Amphetamine 48-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 1831151-9 1991 Both AMPH and stress produced turning, but only AMPH produced widespread c-fos induction, and stress-induced turning only occurred after exposure to AMPH. Amphetamine 48-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 1831151-11 1991 Whereas a high dose of MK-801 (1.0 mg/kg) completely blocked c-fos induction, a lower dose (0.1 mg/kg) blocked c-fos induction in controls, but left patches of fos-immunoreactive neurons in lesioned animals given transplants. Dizocilpine Maleate 23-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 1831151-12 1991 Thus the sensitization of transplant-related behaviors is NMDA dependent and associated with c-fos induction in host striatal neurons. N-Methylaspartate 58-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 1647024-10 1991 & Greenberg, M. E. (1991) Science, in press), who found that depolarization and Ca2+ influx mediate induction of c-fos in PC12 rat pheochromocytoma cells through phosphorylation of CRE-binding protein. Adenosine Monophosphate 1-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 2054934-6 1991 ET-1, TPA, and ionomycin similarly induced the expression of c-fos after 30 minutes, which returned to an undetectable level after 6 hours. Tetradecanoylphorbol Acetate 6-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 2054934-6 1991 ET-1, TPA, and ionomycin similarly induced the expression of c-fos after 30 minutes, which returned to an undetectable level after 6 hours. Ionomycin 15-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 12106478-7 1991 Agents affecting the intracellular cyclic AMP level, forskolin and Ro 20-1724, stimulated c-fos mRNA in a strong and transient fashion with a temporal sequence similar to the response to CGRP. Cyclic AMP 35-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 12106478-7 1991 Agents affecting the intracellular cyclic AMP level, forskolin and Ro 20-1724, stimulated c-fos mRNA in a strong and transient fashion with a temporal sequence similar to the response to CGRP. Colforsin 53-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 12106478-8 1991 Further, the phosphodiesterase inhibitor Ro 20-1724 potentiated the action of CGRP on c-fos mRNA induction, suggesting a role for cyclic AMP in the action of CGRP. Cyclic AMP 130-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 1913186-9 1991 MK-801 (3 mg/kg, i.p., 30 min prior to occlusion) completely blocked the early c-fos protein induction in all regions but expression within neurons surrounding the ischemic core was present 1 day after a single injection. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 1715968-8 1991 The N-methyl-D-aspartate (NMDA) receptor antagonist MK-801 prevented the injury-induced increases in both preprocholecystokinin and c-fos gene expression, indicating that stimulation of this glutamate receptor subtype may initiate the changes in expression of both genes. Dizocilpine Maleate 52-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 1645738-5 1991 RA also caused a rapid increase in the expression of the early response gene, c-fos, but did not effect the expression of egr-1. Tretinoin 0-2 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 1922926-0 1991 The immediate-early genes c-fos and c-jun are differentially expressed in the rat adrenal gland after capsaicin treatment. Capsaicin 102-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 1922926-1 1991 In situ hybridization histochemistry was used to study the expression of c-fos and c-jun mRNA in the rat adrenal gland of untreated and capsaicin treated rats. Capsaicin 136-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 1922926-4 1991 After capsaicin (25 mg/kg, s.c.), a rapid increase in both c-fos and c-jun mRNA levels was observed in adrenal medulla. Capsaicin 6-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 1922926-5 1991 Capsaicin also induced an increase in c-fos mRNA levels in all 3 cortical layers, especially in the zona glomerulosa, whereas only small changes in c-jun mRNA levels were seen in zona fasciculata and reticulata. Capsaicin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 1922926-6 1991 The present results indicate that c-fos and c-jun mRNA levels are both increased in the adrenal gland after capsaicin treatment, although the time course, magnitude and regional distribution of these increases differed for the two mRNAs. Capsaicin 108-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 1845539-0 1991 Acute administration of ethanol suppresses pentylenetetrazole-induced c-fos expression in rat brain. Ethanol 24-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 1828153-7 1991 Ang II (10(9) - 10(-8) M) and a protein kinase C activator, phorbol 12-myristate 13-acetate (PMA, 10(-8) M) rapidly induced c-fos as well as c-Jun and Jun-B mRNA expression in RASM cells. Tetradecanoylphorbol Acetate 60-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 1828153-7 1991 Ang II (10(9) - 10(-8) M) and a protein kinase C activator, phorbol 12-myristate 13-acetate (PMA, 10(-8) M) rapidly induced c-fos as well as c-Jun and Jun-B mRNA expression in RASM cells. Tetradecanoylphorbol Acetate 93-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 1905596-7 1991 These data suggest that the binding of the antibodies to the TSH-receptor followed by cAMP production is related to the induction of c-fos and c-myc mRNAs and, thus, to the growth of FRTL5 cells. Cyclic AMP 86-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 1710164-1 1991 An increase of proto-oncogene c-fos expression in cerebral cortex of rats treated with subconvulsant doses of the pesticide organochlorine lindane (gamma-hexachlorocyclohexane) has been detected using Northern blots. Hexachlorocyclohexane 148-175 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 1710164-7 1991 The co-induction of c-fos and ODC suggests a potential link between the ODC/polyamine system and the short-acting proto-oncogenes in stimulus-transcription coupling events. Polyamines 76-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 1677338-0 1991 3,4-Methylenedioxymethamphetamine induces Fos-like proteins in rat basal ganglia: reversal with MK 801. N-Methyl-3,4-methylenedioxyamphetamine 0-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 1677338-0 1991 3,4-Methylenedioxymethamphetamine induces Fos-like proteins in rat basal ganglia: reversal with MK 801. Dizocilpine Maleate 96-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 1677338-4 1991 The NMDA antagonist MK 801 inhibited Fos and Fos-related antigen induction after MDMA injections, whereas fluoxetine, a serotonin uptake inhibitor, had no effect. N-Methylaspartate 4-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 1677338-4 1991 The NMDA antagonist MK 801 inhibited Fos and Fos-related antigen induction after MDMA injections, whereas fluoxetine, a serotonin uptake inhibitor, had no effect. Dizocilpine Maleate 20-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 1677338-4 1991 The NMDA antagonist MK 801 inhibited Fos and Fos-related antigen induction after MDMA injections, whereas fluoxetine, a serotonin uptake inhibitor, had no effect. N-Methyl-3,4-methylenedioxyamphetamine 81-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 1677338-5 1991 Thus, MDMA induces Fos and Fos-related antigens in striatal neurons in an NMDA-reversible fashion. N-Methyl-3,4-methylenedioxyamphetamine 6-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 1677338-5 1991 Thus, MDMA induces Fos and Fos-related antigens in striatal neurons in an NMDA-reversible fashion. N-Methyl-3,4-methylenedioxyamphetamine 6-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 1677338-5 1991 Thus, MDMA induces Fos and Fos-related antigens in striatal neurons in an NMDA-reversible fashion. N-Methylaspartate 74-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 1677338-5 1991 Thus, MDMA induces Fos and Fos-related antigens in striatal neurons in an NMDA-reversible fashion. N-Methylaspartate 74-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 1902094-1 1991 It was previously shown that injection of 17 beta-estradiol into adult ovariectomized rats induces a rapid and transient increase of c-fos gene transcription in the uterus. Estradiol 42-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 1908674-2 1991 A single injection of norepinephrine (2.5 micrograms/kg to 2.5 mg/kg) led to transient increases in the levels of both c-fos and c-myc mRNA. Norepinephrine 22-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 1899867-4 1991 Stimulation with arginine vasopressin, epidermal growth factor, or phorbol myristate acetate increased [3H]thymidine incorporation and mRNA levels of the immediate-early response genes c-fos and Egr-1. Tetradecanoylphorbol Acetate 67-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 185-190 1899867-4 1991 Stimulation with arginine vasopressin, epidermal growth factor, or phorbol myristate acetate increased [3H]thymidine incorporation and mRNA levels of the immediate-early response genes c-fos and Egr-1. Tritium 104-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 185-190 1899867-4 1991 Stimulation with arginine vasopressin, epidermal growth factor, or phorbol myristate acetate increased [3H]thymidine incorporation and mRNA levels of the immediate-early response genes c-fos and Egr-1. Thymidine 107-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 185-190 1702436-5 1991 The transfected chloramphenicol acetyltransferase gene linked to upstream sequences of the fos gene indicated that sequences containing a serum response element were required for efficient transcription by stretching and that sequences containing a cAMP/calcium response element might not be involved in the c-fos response to myocyte stretching. Cyclic AMP 249-253 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-94 1702436-5 1991 The transfected chloramphenicol acetyltransferase gene linked to upstream sequences of the fos gene indicated that sequences containing a serum response element were required for efficient transcription by stretching and that sequences containing a cAMP/calcium response element might not be involved in the c-fos response to myocyte stretching. Calcium 254-261 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-94 1702436-6 1991 The accumulation of c-fos mRNA by stretching was suppressed by protein kinase C inhibitors at the transcriptional level and inhibited markedly by down-regulation of protein kinase C. Moreover, myocyte stretching increased inositol phosphate levels, and activation of protein kinase C by phorbol esters stimulated the expression of c-fos and skeletal alpha-actin genes. Inositol Phosphates 222-240 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 1702436-6 1991 The accumulation of c-fos mRNA by stretching was suppressed by protein kinase C inhibitors at the transcriptional level and inhibited markedly by down-regulation of protein kinase C. Moreover, myocyte stretching increased inositol phosphate levels, and activation of protein kinase C by phorbol esters stimulated the expression of c-fos and skeletal alpha-actin genes. Phorbol Esters 287-301 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 1662302-2 1991 The c-fos-immunopositive neurons were observed in a hippocampal formation, especially in the dentate gyrus and CA1 and CA2, in the vicinity of the iron injected cerebral cortex and amygdala only at 3 hours after the iron administration. Iron 147-151 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 1662302-2 1991 The c-fos-immunopositive neurons were observed in a hippocampal formation, especially in the dentate gyrus and CA1 and CA2, in the vicinity of the iron injected cerebral cortex and amygdala only at 3 hours after the iron administration. Iron 216-220 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 1901946-8 1991 The same alkaline phosphatase treatment completely abolished the DNA-binding properties of the c-fos cyclic AMP-responsive element-specific proteins. Cyclic AMP 101-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 1903567-0 1991 Inhibition of the 17 beta-estradiol-induced and constitutive expression of the cellular protooncogene c-fos by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in the female rat uterus. Estradiol 18-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 1903567-0 1991 Inhibition of the 17 beta-estradiol-induced and constitutive expression of the cellular protooncogene c-fos by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in the female rat uterus. Polychlorinated Dibenzodioxins 111-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 1903567-0 1991 Inhibition of the 17 beta-estradiol-induced and constitutive expression of the cellular protooncogene c-fos by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in the female rat uterus. Polychlorinated Dibenzodioxins 148-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 1903567-1 1991 Acute administration of 17 beta-estradiol (5 micrograms/rat) to 25-day-old female Sprague-Dawley rats resulted in an increase of uterine mRNA for the cellular oncogene c-fos. Estradiol 24-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-173 1903567-2 1991 The c-fos mRNA levels were significantly elevated 12 and 24 h after exposure to the hormone (232 and 164% of control values) and the elevation was not observed after 48 h. In contrast, treatment of the animals with either 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) or 6-methyl-1,3,8-trichlorodibenzofuran (MCDF) resulted in a dose-dependent decrease in constitutive uterine c-fos mRNA levels. Polychlorinated Dibenzodioxins 259-263 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 1903567-2 1991 The c-fos mRNA levels were significantly elevated 12 and 24 h after exposure to the hormone (232 and 164% of control values) and the elevation was not observed after 48 h. In contrast, treatment of the animals with either 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) or 6-methyl-1,3,8-trichlorodibenzofuran (MCDF) resulted in a dose-dependent decrease in constitutive uterine c-fos mRNA levels. 6-methyl-1,3,8-trichlorodibenzofuran 268-304 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 1903567-2 1991 The c-fos mRNA levels were significantly elevated 12 and 24 h after exposure to the hormone (232 and 164% of control values) and the elevation was not observed after 48 h. In contrast, treatment of the animals with either 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) or 6-methyl-1,3,8-trichlorodibenzofuran (MCDF) resulted in a dose-dependent decrease in constitutive uterine c-fos mRNA levels. 6-methyl-1,3,8-trichlorodibenzofuran 306-310 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 1903567-3 1991 In rats co-treated with 17 beta-estradiol plus TCDD or MCDF, it was apparent from the results that the halogenated aromatic hydrocarbons significantly inhibited the estrogen-induced increases in uterine c-fos mRNA levels. Estradiol 24-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 203-208 1903567-3 1991 In rats co-treated with 17 beta-estradiol plus TCDD or MCDF, it was apparent from the results that the halogenated aromatic hydrocarbons significantly inhibited the estrogen-induced increases in uterine c-fos mRNA levels. Polychlorinated Dibenzodioxins 47-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 203-208 1903567-3 1991 In rats co-treated with 17 beta-estradiol plus TCDD or MCDF, it was apparent from the results that the halogenated aromatic hydrocarbons significantly inhibited the estrogen-induced increases in uterine c-fos mRNA levels. 6-methyl-1,3,8-trichlorodibenzofuran 55-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 203-208 1903567-3 1991 In rats co-treated with 17 beta-estradiol plus TCDD or MCDF, it was apparent from the results that the halogenated aromatic hydrocarbons significantly inhibited the estrogen-induced increases in uterine c-fos mRNA levels. Hydrocarbons, Aromatic 115-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 203-208 1903567-4 1991 These observations further extend the diverse spectrum of antiestrogenic effects caused by TCDD and related compounds and also show an interaction between TCDD and the constitutive expression of the c-fos proto-oncogene in the female rat uterus. Polychlorinated Dibenzodioxins 91-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-204 1903567-4 1991 These observations further extend the diverse spectrum of antiestrogenic effects caused by TCDD and related compounds and also show an interaction between TCDD and the constitutive expression of the c-fos proto-oncogene in the female rat uterus. Polychlorinated Dibenzodioxins 155-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-204 1710164-1 1991 An increase of proto-oncogene c-fos expression in cerebral cortex of rats treated with subconvulsant doses of the pesticide organochlorine lindane (gamma-hexachlorocyclohexane) has been detected using Northern blots. organochlorine lindane 124-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 1825356-0 1991 Cocaine induces striatal c-fos-immunoreactive proteins via dopaminergic D1 receptors. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 1825356-3 1991 We report here that the acute administration of a single dose of the indirect-acting dopaminergic agonist cocaine increases multiple Fos proteins in rat caudate nucleus. Cocaine 106-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-136 1845539-0 1991 Acute administration of ethanol suppresses pentylenetetrazole-induced c-fos expression in rat brain. Pentylenetetrazole 43-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 1845539-1 1991 The effect of acute ethanol administration on pentylenetetrazole-induced c-fos expression in rat brain was studied. Ethanol 20-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 1715678-0 1991 Striatal c-fos induction in neonatally dopamine-depleted rats given transplants. Dopamine 39-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 1845539-1 1991 The effect of acute ethanol administration on pentylenetetrazole-induced c-fos expression in rat brain was studied. Pentylenetetrazole 46-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 1845539-2 1991 Pentylenetetrazole induced the rapid and transient expression of c-fos mRNA in rat brain. Pentylenetetrazole 0-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 1845575-7 1991 A distinct increase in expression of c-fos gene in brain cortex was observed in offsprings of chronically alcohol-treated rats after administration of ethanol. Alcohols 106-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 1845575-7 1991 A distinct increase in expression of c-fos gene in brain cortex was observed in offsprings of chronically alcohol-treated rats after administration of ethanol. Ethanol 151-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 1845575-8 1991 The impairments in regulation of c-fos gene activity in offsprings suggest that chronic alcohol treatment of animals may influence functions of genome of their offsprings which were not subjected to chronic alcohol treatment. Alcohols 88-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 1845575-8 1991 The impairments in regulation of c-fos gene activity in offsprings suggest that chronic alcohol treatment of animals may influence functions of genome of their offsprings which were not subjected to chronic alcohol treatment. Alcohols 207-214 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 1687822-5 1991 Other gene, cfos, is also induced by reserpine indicating that the TH transcription in these neurons may be mediated by "third messengers". Reserpine 37-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-16 1773755-5 1991 In contrast to the diffuse and bilateral cortical staining observed with electrical stimulation, motor cortex stimulation by the local application of bicuculline resulted in c-fos protein staining restricted to the stimulated motor cortex. Bicuculline 150-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 174-179 1680735-2 1991 Induction of modular patterns of fos-like immunoreactivity by cocaine. Cocaine 62-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 1680735-3 1991 Cocaine, a catecholamine agonist, has been shown to produce a transient induction of the immediate-early gene c-fos and its protein product Fos in the striatum of normal rats. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 1680735-3 1991 Cocaine, a catecholamine agonist, has been shown to produce a transient induction of the immediate-early gene c-fos and its protein product Fos in the striatum of normal rats. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-143 1680735-3 1991 Cocaine, a catecholamine agonist, has been shown to produce a transient induction of the immediate-early gene c-fos and its protein product Fos in the striatum of normal rats. Catecholamines 11-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 1680735-3 1991 Cocaine, a catecholamine agonist, has been shown to produce a transient induction of the immediate-early gene c-fos and its protein product Fos in the striatum of normal rats. Catecholamines 11-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-143 1680735-4 1991 In the present study we report that the expression of Fos can be induced by cocaine challenge in intrastriatal grafts derived from cell suspensions of embryonic striatal primordia. Cocaine 76-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 1680735-5 1991 Fos-like immunoreactivity in the nuclei of grafted neurons was detected 2 hr after the injection of 50 mg/kg cocaine into the host rats. Cocaine 109-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 1680735-9 1991 The preferential expression of Fos in neurons of the P regions of the grafts thus implies that the induction of Fos was cell-type specific in being concentrated in the parts of the grafts that express striatal phenotype and that are innervated by catecholamine-containing fibers. Catecholamines 247-260 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 1680735-9 1991 The preferential expression of Fos in neurons of the P regions of the grafts thus implies that the induction of Fos was cell-type specific in being concentrated in the parts of the grafts that express striatal phenotype and that are innervated by catecholamine-containing fibers. Catecholamines 247-260 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 12106477-0 1991 Nerve Growth Factor is Required for Induction of c-Fos Immunoreactivity by Serum, Depolarization, Cyclic AMP or Trauma in Cultured Rat Sympathetic Neurons. Cyclic AMP 98-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 1674966-0 1991 Differential effect of lithium on fos protooncogene expression mediated by receptor and postreceptor activators of protein kinase C and cyclic adenosine monophosphate: model for its antimanic action. Lithium 23-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 1674966-0 1991 Differential effect of lithium on fos protooncogene expression mediated by receptor and postreceptor activators of protein kinase C and cyclic adenosine monophosphate: model for its antimanic action. Cyclic AMP 136-166 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 1674966-4 1991 We find that lithium, at therapeutic doses, has an augmenting effect on phosphatidylinositol (PI)-mediated fos expression induced by activating a muscarinic cholinergic pathway, whereas it has no effect, at tenfold the therapeutic dose, on fos expression induced by receptor or postreceptor activators of cyclic adenosine monophosphate (cAMP). Lithium 13-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 1674966-4 1991 We find that lithium, at therapeutic doses, has an augmenting effect on phosphatidylinositol (PI)-mediated fos expression induced by activating a muscarinic cholinergic pathway, whereas it has no effect, at tenfold the therapeutic dose, on fos expression induced by receptor or postreceptor activators of cyclic adenosine monophosphate (cAMP). Lithium 13-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 240-243 1674966-4 1991 We find that lithium, at therapeutic doses, has an augmenting effect on phosphatidylinositol (PI)-mediated fos expression induced by activating a muscarinic cholinergic pathway, whereas it has no effect, at tenfold the therapeutic dose, on fos expression induced by receptor or postreceptor activators of cyclic adenosine monophosphate (cAMP). Phosphatidylinositols 72-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 1674966-4 1991 We find that lithium, at therapeutic doses, has an augmenting effect on phosphatidylinositol (PI)-mediated fos expression induced by activating a muscarinic cholinergic pathway, whereas it has no effect, at tenfold the therapeutic dose, on fos expression induced by receptor or postreceptor activators of cyclic adenosine monophosphate (cAMP). Phosphatidylinositols 72-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 240-243 1674966-8 1991 We also show that carbamazepine, another antimanic agent, has an inhibitory effect on cAMP-mediated fos but no effect on the IP pathway. Carbamazepine 18-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-103 1674966-8 1991 We also show that carbamazepine, another antimanic agent, has an inhibitory effect on cAMP-mediated fos but no effect on the IP pathway. Cyclic AMP 86-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-103 1716733-4 1991 It has been found that in E1Aad5 + cHa-ras-transformed cells the expression of c-fos promoter has a constitutive, non-inducible character while in REF cells and cells immortalized by E1Aad5 the fos-promoter can be regulated by serum growth factors, EGF, and TPA. Tetradecanoylphorbol Acetate 258-261 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 1834961-5 1991 After intrastriatal injection of the N-methyl-D-aspartate receptor agonist, quinolinic acid, an increase in c-fos mRNA concentrations was detected using in situ hybridization and Northern blot analysis. Quinolinic Acid 76-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 1834961-8 1991 Our results indicate that N-methyl-D-aspartate receptor activation by quinolinic acid stimulates medium spiny neurons to increase c-Fos expression; to a lesser extent, medium aspiny interneurons and glial cells also respond. Quinolinic Acid 70-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 1908066-8 1991 Only the mustard oil-treated rats had obvious signs of rhinitis and displayed FOS-positive cells in laminae I and II of the subnucleus caudalis and in the subnucleus interpolaris of the trigeminal brainstem nuclear complex as well as in the medullary lateral reticular nucleus. mustard oil 9-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 1659673-1 1991 In an earlier report, we demonstrated that subcutaneous injection of formalin in the rat hindpaw evokes a characteristic pattern of expression of the fos protein product of the c-fos protooncogene in spinal cord neurons, and that systemic morphine reversed the fos-like immunoreactivity in a dose-dependent, naloxone-reversible manner. Formaldehyde 69-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-153 1659673-1 1991 In an earlier report, we demonstrated that subcutaneous injection of formalin in the rat hindpaw evokes a characteristic pattern of expression of the fos protein product of the c-fos protooncogene in spinal cord neurons, and that systemic morphine reversed the fos-like immunoreactivity in a dose-dependent, naloxone-reversible manner. Formaldehyde 69-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 177-182 1659673-1 1991 In an earlier report, we demonstrated that subcutaneous injection of formalin in the rat hindpaw evokes a characteristic pattern of expression of the fos protein product of the c-fos protooncogene in spinal cord neurons, and that systemic morphine reversed the fos-like immunoreactivity in a dose-dependent, naloxone-reversible manner. Formaldehyde 69-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-182 1659673-1 1991 In an earlier report, we demonstrated that subcutaneous injection of formalin in the rat hindpaw evokes a characteristic pattern of expression of the fos protein product of the c-fos protooncogene in spinal cord neurons, and that systemic morphine reversed the fos-like immunoreactivity in a dose-dependent, naloxone-reversible manner. Morphine 239-247 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 177-182 1659673-1 1991 In an earlier report, we demonstrated that subcutaneous injection of formalin in the rat hindpaw evokes a characteristic pattern of expression of the fos protein product of the c-fos protooncogene in spinal cord neurons, and that systemic morphine reversed the fos-like immunoreactivity in a dose-dependent, naloxone-reversible manner. Naloxone 308-316 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-153 1659673-1 1991 In an earlier report, we demonstrated that subcutaneous injection of formalin in the rat hindpaw evokes a characteristic pattern of expression of the fos protein product of the c-fos protooncogene in spinal cord neurons, and that systemic morphine reversed the fos-like immunoreactivity in a dose-dependent, naloxone-reversible manner. Naloxone 308-316 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 177-182 1659673-1 1991 In an earlier report, we demonstrated that subcutaneous injection of formalin in the rat hindpaw evokes a characteristic pattern of expression of the fos protein product of the c-fos protooncogene in spinal cord neurons, and that systemic morphine reversed the fos-like immunoreactivity in a dose-dependent, naloxone-reversible manner. Naloxone 308-316 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-182 1659673-3 1991 Formalin injection produced a biphasic pain behavioral response which lasted about 1 h. There was a significant correlation between the formalin pain score and overall fos-like immunoreactivity in the lumbar enlargement. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-171 1659673-3 1991 Formalin injection produced a biphasic pain behavioral response which lasted about 1 h. There was a significant correlation between the formalin pain score and overall fos-like immunoreactivity in the lumbar enlargement. Formaldehyde 136-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-171 1659673-4 1991 The greatest numbers of labeled cells and most intense fos-like immunoreactivity were found in laminae I, IIo and V of the L4-5 segments, ipsilateral to the formalin-injected paw. Formaldehyde 157-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 1659673-6 1991 [D-Ala2, NMe-Phe4, Gly-ol5]enkephalin produced a dose-related, naloxone-reversible inhibition of both the formalin-evoked pain behavior and fos expression in the cord. nme-phe4 9-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-143 1659673-6 1991 [D-Ala2, NMe-Phe4, Gly-ol5]enkephalin produced a dose-related, naloxone-reversible inhibition of both the formalin-evoked pain behavior and fos expression in the cord. Naloxone 63-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-143 1659673-10 1991 Furthermore, the dose of [D-Ala2, NMe-Phe4, Gly-ol5]enkephalin which produced approximately 50% inhibition of fos-like immunoreactivity in the neck and ventral regions of the spinal cord was without effect in the superficial dorsal horn. nme-phe4 34-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 1659673-10 1991 Furthermore, the dose of [D-Ala2, NMe-Phe4, Gly-ol5]enkephalin which produced approximately 50% inhibition of fos-like immunoreactivity in the neck and ventral regions of the spinal cord was without effect in the superficial dorsal horn. gly-ol5 44-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 1659673-12 1991 Finally, we found that bilateral, midthoracic lesions of the dorsal part of the lateral funiculus blocked both the antinociception and fos suppression produced by intracerebroventricular [D-Ala2, NMe-Phe4, Gly-ol5]enkephalin. nme-phe4 196-204 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-138 2128198-0 1990 Effects of kainic acid-induced seizures and ischemia on c-fos-like proteins in rat brain. Kainic Acid 11-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 2128198-1 1990 We have analyzed the brain pattern and time-course of c-fos-like proteins expression in kainic acid-induced seizures in the rat. Kainic Acid 88-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 2128198-5 1990 This time-course is similar to that produced by kainic acid on 2-deoxyglucose consumption and correlates with the electrographic changes previously described, supporting the idea that c-fos-like immunostaining may provide a useful marker of neuronal activity, with a cellular resolution. Kainic Acid 48-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 184-189 2127308-0 1990 Acute administration of ethanol suppresses pentylenetetrazole-induced c-fos expression in rat brain. Ethanol 24-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 2127308-0 1990 Acute administration of ethanol suppresses pentylenetetrazole-induced c-fos expression in rat brain. Pentylenetetrazole 43-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 2127308-1 1990 The effect of acute ethanol administration on pentylenetetrazole-induced c-fos expression in rat brain was studied. Ethanol 20-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 2127308-1 1990 The effect of acute ethanol administration on pentylenetetrazole-induced c-fos expression in rat brain was studied. Pentylenetetrazole 46-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 2127308-2 1990 Pentylenetetrazole induced the rapid and transient expression of c-fos mRNA in rat brain. Pentylenetetrazole 0-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 2178237-2 1990 Within minutes of addition, agents such as nerve growth factor (NGF), epidermal growth factor (EGF), basic fibroblast growth factor (bFGF) and dibutyryl cyclic AMP (db cAMP) rapidly activate cellular immediate early genes such as c-fos, c-jun, jun-B, and egr-1. Bucladesine 143-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 230-235 19912775-7 1990 Since many studies have shown that neuronal development within sexually dimorphic areas of the hypothalamus and spinal cord is influenced by gonadal steroids around the time of birth, we hypothesized that Fos expression within such areas might differ between males and females at birth. Steroids 149-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 205-208 2289147-4 1990 Furthermore, Fos B-like immunoreactivity was induced in neurons by two treatments (focal brain injury and haloperidol injection) that are known to induce Fos, however, whereas Fos levels returned to baseline 24 h after these treatments, Fos B-like immunoreactivity remained elevated at this time point. Haloperidol 106-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 2289147-4 1990 Furthermore, Fos B-like immunoreactivity was induced in neurons by two treatments (focal brain injury and haloperidol injection) that are known to induce Fos, however, whereas Fos levels returned to baseline 24 h after these treatments, Fos B-like immunoreactivity remained elevated at this time point. Haloperidol 106-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-157 2289147-4 1990 Furthermore, Fos B-like immunoreactivity was induced in neurons by two treatments (focal brain injury and haloperidol injection) that are known to induce Fos, however, whereas Fos levels returned to baseline 24 h after these treatments, Fos B-like immunoreactivity remained elevated at this time point. Haloperidol 106-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-157 2289147-4 1990 Furthermore, Fos B-like immunoreactivity was induced in neurons by two treatments (focal brain injury and haloperidol injection) that are known to induce Fos, however, whereas Fos levels returned to baseline 24 h after these treatments, Fos B-like immunoreactivity remained elevated at this time point. Haloperidol 106-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-157 2289147-5 1990 Also, although focal brain injury and rolipram injections induced Fos in ependymal and glial-like cells in rat brain, Fos B-like immunoreactivity was not detected in these non-neural cells. Rolipram 38-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 2289147-5 1990 Also, although focal brain injury and rolipram injections induced Fos in ependymal and glial-like cells in rat brain, Fos B-like immunoreactivity was not detected in these non-neural cells. Rolipram 38-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 2124525-0 1990 Expression of c-fos protein by N-methyl-D-aspartic acid in hypothalamus of immature female rats: blockade by MK-801 or neonatal treatment with monosodium glutamate. N-Methylaspartate 31-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 2124525-0 1990 Expression of c-fos protein by N-methyl-D-aspartic acid in hypothalamus of immature female rats: blockade by MK-801 or neonatal treatment with monosodium glutamate. Dizocilpine Maleate 109-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 2124525-0 1990 Expression of c-fos protein by N-methyl-D-aspartic acid in hypothalamus of immature female rats: blockade by MK-801 or neonatal treatment with monosodium glutamate. Sodium Glutamate 143-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 2124525-1 1990 Significant NMDA-induced cellular activity in the median eminence-arcuate region of immature female rats was immunocytochemically localized via c-fos protein-like immunoreactivity. N-Methylaspartate 12-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-149 2124525-3 1990 Treatment of newborn rats with monosodium glutamate (MSG), which destroys glutamate-sensitive neurons, significantly attenuated NMDA-induced c-fos when tested in the peripubertal period. Sodium Glutamate 31-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 2124525-3 1990 Treatment of newborn rats with monosodium glutamate (MSG), which destroys glutamate-sensitive neurons, significantly attenuated NMDA-induced c-fos when tested in the peripubertal period. Sodium Glutamate 53-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 2124525-3 1990 Treatment of newborn rats with monosodium glutamate (MSG), which destroys glutamate-sensitive neurons, significantly attenuated NMDA-induced c-fos when tested in the peripubertal period. Glutamic Acid 42-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 2124525-3 1990 Treatment of newborn rats with monosodium glutamate (MSG), which destroys glutamate-sensitive neurons, significantly attenuated NMDA-induced c-fos when tested in the peripubertal period. N-Methylaspartate 128-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 2121467-3 1990 RU486 treatment greatly reduced both the magnitude of the LH surge and the degree of cFos induction (numbers of cells expressing cFos and intensity of cFos staining) in LHRH neurons during proestrus. Mifepristone 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-89 2121467-3 1990 RU486 treatment greatly reduced both the magnitude of the LH surge and the degree of cFos induction (numbers of cells expressing cFos and intensity of cFos staining) in LHRH neurons during proestrus. Mifepristone 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-133 2121467-3 1990 RU486 treatment greatly reduced both the magnitude of the LH surge and the degree of cFos induction (numbers of cells expressing cFos and intensity of cFos staining) in LHRH neurons during proestrus. Mifepristone 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-133 2121467-5 1990 Treatment with EB without P resulted in significantly lower peak LH levels and a reduced cFos response in LHRH neurons than the EB-P treated rats. estradiol 3-benzoate 15-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-93 2126969-6 1990 MK-801 or carbamazepine, which prevented hypoxia-ischemia-induced seizures, also prevented c-fos induction in the non-ligated hemisphere while MK-801 was associated with increased c-fos induction in hippocampal neurones from the ligated side, as well as in glial-like and ependymal cells. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 2126969-6 1990 MK-801 or carbamazepine, which prevented hypoxia-ischemia-induced seizures, also prevented c-fos induction in the non-ligated hemisphere while MK-801 was associated with increased c-fos induction in hippocampal neurones from the ligated side, as well as in glial-like and ependymal cells. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 180-185 2126969-6 1990 MK-801 or carbamazepine, which prevented hypoxia-ischemia-induced seizures, also prevented c-fos induction in the non-ligated hemisphere while MK-801 was associated with increased c-fos induction in hippocampal neurones from the ligated side, as well as in glial-like and ependymal cells. Carbamazepine 10-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 2126969-6 1990 MK-801 or carbamazepine, which prevented hypoxia-ischemia-induced seizures, also prevented c-fos induction in the non-ligated hemisphere while MK-801 was associated with increased c-fos induction in hippocampal neurones from the ligated side, as well as in glial-like and ependymal cells. Carbamazepine 10-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 180-185 2126969-6 1990 MK-801 or carbamazepine, which prevented hypoxia-ischemia-induced seizures, also prevented c-fos induction in the non-ligated hemisphere while MK-801 was associated with increased c-fos induction in hippocampal neurones from the ligated side, as well as in glial-like and ependymal cells. Dizocilpine Maleate 143-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 180-185 2126973-2 1990 C-fos immunoreactivity was detected in a subset of OXY neurons, as well as in other neurons non-immunoreactive for OXY, as early as 90 min after intraperitoneal injection of a hypertonic saline solution. Sodium Chloride 187-193 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 2221010-3 1990 mRNA levels of c-fos and c-jun from aortic smooth muscle cells (SMC) were simultaneously induced within 30 minutes of BDE and declined to baseline by 1.5 hours, c-myc mRNA did not begin to increase until 1 hour after vascular injury. 3,4-epoxy-1,2,3,4-tetrahydrobenzene-1,2-diol 118-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 2176705-4 1990 Sodium pentobarbital, which blocks the induction of Fos-like immunoreactivity (Fos-IR), also blocked a long-duration form of LTP maintained over weeks. Pentobarbital 0-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 2176705-4 1990 Sodium pentobarbital, which blocks the induction of Fos-like immunoreactivity (Fos-IR), also blocked a long-duration form of LTP maintained over weeks. Pentobarbital 0-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 19912764-4 1990 The decline in the number of c-fos-immunoreactive neurons in the Ammon"s horn was partially reversed by a single intravenous injection of estradiol which resulted in the expression of c-fos in 71% of the neurons in area CA(1) and 74% in CA(3) when compared to the numbers of positive cells in the control animals. Estradiol 138-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 19912764-4 1990 The decline in the number of c-fos-immunoreactive neurons in the Ammon"s horn was partially reversed by a single intravenous injection of estradiol which resulted in the expression of c-fos in 71% of the neurons in area CA(1) and 74% in CA(3) when compared to the numbers of positive cells in the control animals. Estradiol 138-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 184-189 1660860-7 1991 Similarly, in cultures incubated with 8-bromo-cAMP or isoprenaline, more than 70% of the pinealocytes were Fos positive. 8-Bromo Cyclic Adenosine Monophosphate 38-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 1660860-7 1991 Similarly, in cultures incubated with 8-bromo-cAMP or isoprenaline, more than 70% of the pinealocytes were Fos positive. Isoproterenol 54-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 1660860-8 1991 The c-fos gene could be involved in the regulation of pineal melatonin synthesis and SRE and Ca/CRE probably participate in its activation process in pinealocytes. Melatonin 61-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 1698651-1 1990 Cortical cavity lesions and lateral ventricular injections of quinolinic acid, a NMDA receptor agonist, induce Fos and Fos-related antigens (FRAs) throughout ipsilateral adult rat brain cortex in similar patterns. Quinolinic Acid 62-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 1698651-1 1990 Cortical cavity lesions and lateral ventricular injections of quinolinic acid, a NMDA receptor agonist, induce Fos and Fos-related antigens (FRAs) throughout ipsilateral adult rat brain cortex in similar patterns. Quinolinic Acid 62-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-122 1698651-6 1990 Intraventricular injections of quisqualate, a non-NMDA glutamate analogue, induced Fos in hippocampus but not in cortex. Quisqualic Acid 31-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 2127073-1 1990 Antibodies to c-fos oncoprotein were produced in rabbits by immunization with synthetic peptides, corresponding to the sequences 6-15 of N-end and 371-380 of C-end of c-fos oncoprotein. Peptides 88-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 2127073-1 1990 Antibodies to c-fos oncoprotein were produced in rabbits by immunization with synthetic peptides, corresponding to the sequences 6-15 of N-end and 371-380 of C-end of c-fos oncoprotein. Peptides 88-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 167-172 1696944-1 1990 The effect of cadmium on the expression of oncogenes c-jun, c-fos, and c-myc was studied by exposing L6J1 myoblast cultures to different doses of cadmium chloride and then analyzing the abundance of oncogene transcripts. Cadmium 14-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 1696944-2 1990 Cadmium induced a transient accumulation of c-jun and c-myc mRNA with maximum expression at 2-4 h. At the same time, the level of c-fos transcripts remained below the detection level. Cadmium 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 1696944-8 1990 Induction of c-myc and c-jun by cadmium and c-fos by a combination of cadmium and cycloheximide could be abolished by blocking transcription with actinomycin D. Cadmium 70-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 1696944-8 1990 Induction of c-myc and c-jun by cadmium and c-fos by a combination of cadmium and cycloheximide could be abolished by blocking transcription with actinomycin D. Cycloheximide 82-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 1696944-8 1990 Induction of c-myc and c-jun by cadmium and c-fos by a combination of cadmium and cycloheximide could be abolished by blocking transcription with actinomycin D. Dactinomycin 146-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 1701330-0 1990 Induction of the c-fos proto-oncogene during opiate withdrawal in the locus coeruleus and other regions of rat brain. Opiate Alkaloids 45-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 1701330-2 1990 Precipitation of opiate withdrawal in rats, which is known to increase LC firing rates 4-fold, led to a two- to three-fold increase in levels of mRNA and protein for c-fos in the LC 1-2 h after initiation of withdrawal. Opiate Alkaloids 17-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-171 1701330-3 1990 In contrast, levels of c-fos expression were decreased in LC from rats treated acutely or chronically with morphine but not experiencing withdrawal, conditions under which LC firing rates are depressed. Morphine 107-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 1701330-4 1990 Similar regulation of c-fos expression during opiate withdrawal was found in the amygdala, ventral tegmentum, nucleus accumbens, neostriatum, and cerebral cortex, but not in a number of other brain regions studied, which included the hippocampus, dorsal raphe, periaqueductal gray, and paragigantocellularis. Opiate Alkaloids 46-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 1701330-6 1990 The results demonstrate a novel use of c-fos in neuropharmacology, namely to map neuronal pathways and neuronal cell types activated in response to acute and chronic opiate administration and during opiate withdrawal, as well as in response to other psychotropic drug treatments. Opiate Alkaloids 166-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 2166040-5 1990 Vanadate was not a general transcription inhibitor since, it like insulin, stimulated the expression of the c-fos gene. Vanadates 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 2272313-5 1990 These effects of retinoic acid were preceded by an increased expression of proto-oncogene raf and transient expression of proto-oncogene fos. Tretinoin 17-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-140 2272313-6 1990 The maximum level of fos expression was observed at 15 min and of raf at 12 hr after exposure to retinoic acid. Tretinoin 97-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 2178237-5 1990 Transcriptional activation of c-fos, c-jun, jun-B, and egr-1 by NGF, EGF, and db cAMP was down-regulated to a varying extent whereas NGF-induced ornithine decarboxylase (ODC) was not affected. Cyclic AMP 81-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 19912752-1 1990 Effects of gonadal steroids on fos expression in the adult rat ventromedial hypothalamus (VMH) and uterus were examined using molecular hybridization and immunocytochemical techniques. Steroids 19-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 19912752-11 1990 For comparison, intense fos-like IR in the hippocampal formation and in the paraventricular nucleus of the hypothalamus was observed following metrazol-induced seizure activity and water deprivation, respectively. Pentylenetetrazole 143-151 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 19912752-11 1990 For comparison, intense fos-like IR in the hippocampal formation and in the paraventricular nucleus of the hypothalamus was observed following metrazol-induced seizure activity and water deprivation, respectively. Water 181-186 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 2114401-10 1990 Protein synthesis inhibitors (cycloheximide, anisomycin) also induced the transcription of the c-fos gene. Cycloheximide 30-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 2114401-10 1990 Protein synthesis inhibitors (cycloheximide, anisomycin) also induced the transcription of the c-fos gene. Anisomycin 45-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 2114646-5 1990 The timing of onset of Fos expression in LHRH neurons during proestrus suggests a strong correlation with increased LH secretion. Luteinizing Hormone 41-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 2114646-6 1990 Pentobarbital, which blocks the preovulatory LH surge, blocked Fos expression in LHRH neurons, but the LHRH neurons expressed Fos on the following afternoon at the time of the expected delayed LH surge. Pentobarbital 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 2114014-8 1990 The different gel mobility of the Fos immunoreactive bands induced by sodium butyrate and the sustained Fos mRNA levels after induction suggested that the sodium butyrate-induced c-Fos protein could be non-functional in the autoregulation of the c-fos gene. Butyric Acid 70-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-184 2119851-0 1990 Lithium augments fos protoonocogene expression in PC12 pheochromocytoma cells: implications for therapeutic action of lithium. Lithium 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 2119851-4 1990 Because of these interactions, we investigated the effect of lithium on fos gene expression in PC12 pheochromocytoma cells. Lithium 61-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 2119851-5 1990 We find that lithium increases the level of fos mRNA that occurs in response to receptor and postreceptor activation of PKC. Lithium 13-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 2119851-6 1990 Treatment with lithium also leads to an augmentation of muscarinic cholinergic-mediated fos gene expression in cells that are down-regulated as a result of excessive cholinergic stimulation. Lithium 15-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 2114014-8 1990 The different gel mobility of the Fos immunoreactive bands induced by sodium butyrate and the sustained Fos mRNA levels after induction suggested that the sodium butyrate-induced c-Fos protein could be non-functional in the autoregulation of the c-fos gene. Butyric Acid 155-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 2114014-8 1990 The different gel mobility of the Fos immunoreactive bands induced by sodium butyrate and the sustained Fos mRNA levels after induction suggested that the sodium butyrate-induced c-Fos protein could be non-functional in the autoregulation of the c-fos gene. Butyric Acid 155-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 2114014-8 1990 The different gel mobility of the Fos immunoreactive bands induced by sodium butyrate and the sustained Fos mRNA levels after induction suggested that the sodium butyrate-induced c-Fos protein could be non-functional in the autoregulation of the c-fos gene. Butyric Acid 155-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-184 2114014-8 1990 The different gel mobility of the Fos immunoreactive bands induced by sodium butyrate and the sustained Fos mRNA levels after induction suggested that the sodium butyrate-induced c-Fos protein could be non-functional in the autoregulation of the c-fos gene. Butyric Acid 155-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 246-251 2114014-9 1990 Gel shift analysis showed unimpaired capacity of the butyrate-induced c-Fos protein to participate in the formation of transcriptional complexes with the Jun/AP-1 protein. Butyrates 53-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 2114014-10 1990 However, transfection experiments indicate that the sodium butyrate-induced c-Fos protein is not able to negatively trans-regulate the c-fos promoter. Butyric Acid 52-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 2169754-4 1990 Recently we have shown that beta-adrenergic receptor stimulation of rat parotid acinar cells in vitro or addition of 8-BrcAMP in a rat submandibular cell line (RSMT-A5) increases the expression of the c-fos gene in a time-dependent manner. 8-Bromo Cyclic Adenosine Monophosphate 117-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 201-206 2169754-6 1990 The expression of the c-fos gene did not correlate with DNA synthesis in either cell type, and c-fos transcripts were undetectable in the glands of animals treated for eight days with isoproterenol. Isoproterenol 184-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 2115338-7 1990 Both histamine and bFGF induced c-fos gene expression transiently. Histamine 5-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 2166202-2 1990 Marked induction of c-fos mRNA in astrocytes was observed after treatment with epidermal growth factor (EGF), basic fibroblast growth factor (bFGF), dibutyryl cyclic AMP (db-cAMP), and phorbol diester (TPA; 12-O-tetra-decanoylphorbol 13-acetate), which are known to induce the proliferation or differentiation of astrocytes. Bucladesine 149-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 2166202-2 1990 Marked induction of c-fos mRNA in astrocytes was observed after treatment with epidermal growth factor (EGF), basic fibroblast growth factor (bFGF), dibutyryl cyclic AMP (db-cAMP), and phorbol diester (TPA; 12-O-tetra-decanoylphorbol 13-acetate), which are known to induce the proliferation or differentiation of astrocytes. Bucladesine 171-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 2166202-2 1990 Marked induction of c-fos mRNA in astrocytes was observed after treatment with epidermal growth factor (EGF), basic fibroblast growth factor (bFGF), dibutyryl cyclic AMP (db-cAMP), and phorbol diester (TPA; 12-O-tetra-decanoylphorbol 13-acetate), which are known to induce the proliferation or differentiation of astrocytes. Phorbol Esters 185-200 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 2166202-2 1990 Marked induction of c-fos mRNA in astrocytes was observed after treatment with epidermal growth factor (EGF), basic fibroblast growth factor (bFGF), dibutyryl cyclic AMP (db-cAMP), and phorbol diester (TPA; 12-O-tetra-decanoylphorbol 13-acetate), which are known to induce the proliferation or differentiation of astrocytes. Tetradecanoylphorbol Acetate 202-205 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 2166202-3 1990 Increase of c-fos protein immunoreactivity (IR) was obtained after treatment with fetal calf serum, EGF, bFGF, db-cAMP and TPA. Bucladesine 111-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 2166202-3 1990 Increase of c-fos protein immunoreactivity (IR) was obtained after treatment with fetal calf serum, EGF, bFGF, db-cAMP and TPA. Tetradecanoylphorbol Acetate 123-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 2166202-4 1990 High concentrations of calcium ionophore A23187, which were lethal to cultured astrocytes, also increased c-fos protein-IR. Calcium 23-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 2166202-4 1990 High concentrations of calcium ionophore A23187, which were lethal to cultured astrocytes, also increased c-fos protein-IR. Calcimycin 41-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 2114328-5 1990 In the zona granulosa c-fos IR was observed only at 20.00 h and 24.00 h. Administration of dexamethasone, a potent inhibitor of ACTH release, significantly reduced the number of c-fos IR cells. Dexamethasone 91-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 1694182-5 1990 When comparing the subnuclear distribution of the c-fos product with that of densely packed DNA, stained with the fluorochrome Hoechst, an inverse relationship was found. hoechst 127-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 2165580-0 1990 MK801 induces c-fos protein in thalamic and neocortical neurons of rat brain. Dizocilpine Maleate 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 2165580-1 1990 MK801, a non-competitive NMDA receptor antagonist, leads to a dramatic induction of c-fos-like protein in neurons in deep layers of the neocortex, in dorsal and ventral midline thalamic nuclei and in neurons in the central grey of rat brain. Dizocilpine Maleate 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 2165580-2 1990 This induction of c-fos by MK801 is dose- and time-dependent occurring within 2 h and dissipating by 24 h after injection (0.5-8.0mg/kg, i.p.). Dizocilpine Maleate 27-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 2165580-3 1990 The mechanism of this paradoxical induction of c-fos by MK801 is unclear, however the pattern of induction appears to follow the distribution of the antagonist-preferring NMDA receptor site. Dizocilpine Maleate 56-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 2114328-5 1990 In the zona granulosa c-fos IR was observed only at 20.00 h and 24.00 h. Administration of dexamethasone, a potent inhibitor of ACTH release, significantly reduced the number of c-fos IR cells. Dexamethasone 91-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 178-183 2110508-3 1990 When coinjected with an SRE oligonucleotide, it induced c-fos expression in quiescent cells, whereas injection of SRE sequence alone failed to do so. Oligonucleotides 28-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 2109683-11 1990 These data demonstrate that 1) c-fos and c-myc are expressed in ovarian granulosa cells; 2) the expression of the genes encoding c-fos, c-myc, and beta-actin is rapidly increased by gonadotropin; and 3) the increase in the corresponding products of the c-fos and the c-myc genes precedes an increase in DNA synthesis and steroid production. Steroids 321-328 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 2109683-11 1990 These data demonstrate that 1) c-fos and c-myc are expressed in ovarian granulosa cells; 2) the expression of the genes encoding c-fos, c-myc, and beta-actin is rapidly increased by gonadotropin; and 3) the increase in the corresponding products of the c-fos and the c-myc genes precedes an increase in DNA synthesis and steroid production. Steroids 321-328 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 2109683-11 1990 These data demonstrate that 1) c-fos and c-myc are expressed in ovarian granulosa cells; 2) the expression of the genes encoding c-fos, c-myc, and beta-actin is rapidly increased by gonadotropin; and 3) the increase in the corresponding products of the c-fos and the c-myc genes precedes an increase in DNA synthesis and steroid production. Steroids 321-328 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 2108933-0 1990 Enhanced expression of the proto-oncogenes fos and raf in the rhabdomyosarcoma cell line BA-HAN-1C after differentiation induction with retinoic acid and N-methylformamide. Tretinoin 136-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 2108933-0 1990 Enhanced expression of the proto-oncogenes fos and raf in the rhabdomyosarcoma cell line BA-HAN-1C after differentiation induction with retinoic acid and N-methylformamide. methylformamide 154-171 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 2108933-6 1990 Furthermore, a transient expression of c-fos could be observed 15 and 30 min after exposure to RA. Tretinoin 95-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 2099192-0 1990 Heparin inhibits c-fos and c-myc mRNA expression in vascular smooth muscle cells. Heparin 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 2099192-2 1990 In this paper we show that heparin suppressed the induction of c-fos and c-myc mRNA in rat and calf VSMC. Heparin 27-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 2099192-5 1990 The effect of heparin on c-fos mRNA induction was selective for specific mitogens, as heparin inhibited c-fos mRNA induction in phorbol 12-myristate 13-acetate (TPA) stimulated but not epidermal growth factor (EGF) stimulated VSMC. Heparin 14-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 2099192-5 1990 The effect of heparin on c-fos mRNA induction was selective for specific mitogens, as heparin inhibited c-fos mRNA induction in phorbol 12-myristate 13-acetate (TPA) stimulated but not epidermal growth factor (EGF) stimulated VSMC. Heparin 14-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 2099192-5 1990 The effect of heparin on c-fos mRNA induction was selective for specific mitogens, as heparin inhibited c-fos mRNA induction in phorbol 12-myristate 13-acetate (TPA) stimulated but not epidermal growth factor (EGF) stimulated VSMC. Heparin 86-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 2099192-5 1990 The effect of heparin on c-fos mRNA induction was selective for specific mitogens, as heparin inhibited c-fos mRNA induction in phorbol 12-myristate 13-acetate (TPA) stimulated but not epidermal growth factor (EGF) stimulated VSMC. Heparin 86-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 2099192-5 1990 The effect of heparin on c-fos mRNA induction was selective for specific mitogens, as heparin inhibited c-fos mRNA induction in phorbol 12-myristate 13-acetate (TPA) stimulated but not epidermal growth factor (EGF) stimulated VSMC. Tetradecanoylphorbol Acetate 128-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 2099192-5 1990 The effect of heparin on c-fos mRNA induction was selective for specific mitogens, as heparin inhibited c-fos mRNA induction in phorbol 12-myristate 13-acetate (TPA) stimulated but not epidermal growth factor (EGF) stimulated VSMC. Tetradecanoylphorbol Acetate 128-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 2099192-5 1990 The effect of heparin on c-fos mRNA induction was selective for specific mitogens, as heparin inhibited c-fos mRNA induction in phorbol 12-myristate 13-acetate (TPA) stimulated but not epidermal growth factor (EGF) stimulated VSMC. Tetradecanoylphorbol Acetate 161-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 2099192-5 1990 The effect of heparin on c-fos mRNA induction was selective for specific mitogens, as heparin inhibited c-fos mRNA induction in phorbol 12-myristate 13-acetate (TPA) stimulated but not epidermal growth factor (EGF) stimulated VSMC. Tetradecanoylphorbol Acetate 161-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 1690635-0 1990 Regional induction of c-fos-like protein in rat brain after estradiol administration. Estradiol 60-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 1690635-3 1990 After estradiol benzoate (100 micrograms/kg) administration to ovariectomized rats, the number of cells staining for c-fos-like protein increased in the anterior medial preoptic area, the medial preoptic area, the medial amygdala nucleus, and the ventromedial nucleus of hypothalamus, all regions rich in estradiol-concentrating cells. estradiol 3-benzoate 6-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 1690635-3 1990 After estradiol benzoate (100 micrograms/kg) administration to ovariectomized rats, the number of cells staining for c-fos-like protein increased in the anterior medial preoptic area, the medial preoptic area, the medial amygdala nucleus, and the ventromedial nucleus of hypothalamus, all regions rich in estradiol-concentrating cells. Estradiol 6-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 2108173-9 1990 Damage by active oxygen species, pressure overload, and derangements of protein synthesis is likely to include the causative factors of increased expression of c-fos and the hsp 70 gene family in postischemic reperfused liver. Oxygen 17-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 1690271-2 1990 The neuroleptic drug haloperidol, a D2 receptor antagonist, was found to produce a rapid and transient induction of c-fos mRNA expression as compared with the expression in animals treated with saline. Haloperidol 21-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 1690271-3 1990 This induction by haloperidol was found to be dose dependent and D2 receptor mediated, inasmuch as a D2 agonist completely reversed the induction and the inactive isomer of the neuroleptic butaclamol, which does not produce an increase in D2 receptors, had no effect on c-fos mRNA expression. Haloperidol 18-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 270-275 1690271-3 1990 This induction by haloperidol was found to be dose dependent and D2 receptor mediated, inasmuch as a D2 agonist completely reversed the induction and the inactive isomer of the neuroleptic butaclamol, which does not produce an increase in D2 receptors, had no effect on c-fos mRNA expression. Butaclamol 189-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 270-275 2108319-8 1990 Dexamethasone-induced expression of activated N-ras p21 results in blocking of c-fos RNA induction by NGF or bFGF in a time-dependent manner. Dexamethasone 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 2110635-0 1990 MK-801 induces c-fos protein in thalamic and neocortical neurons of rat brain. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 2114014-0 1990 Unusual c-fos induction upon chromaffin PC12 differentiation by sodium butyrate: loss of fos autoregulatory function. chromaffin 29-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-13 2114014-0 1990 Unusual c-fos induction upon chromaffin PC12 differentiation by sodium butyrate: loss of fos autoregulatory function. chromaffin 29-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-13 2114014-0 1990 Unusual c-fos induction upon chromaffin PC12 differentiation by sodium butyrate: loss of fos autoregulatory function. Butyric Acid 64-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-13 2114014-0 1990 Unusual c-fos induction upon chromaffin PC12 differentiation by sodium butyrate: loss of fos autoregulatory function. Butyric Acid 64-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-13 2114014-2 1990 We found that induction of c-fos and c-jun proto-oncogene mRNAs levels following the endocrine differentiation of PC12 cells by sodium butyrate is uncoupled. Butyric Acid 128-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 2114014-4 1990 Continuous presence of sodium butyrate for 72 hours resulted in stable high levels of c-fos and c-jun mRNAs. Butyric Acid 23-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 2114014-8 1990 The different gel mobility of the Fos immunoreactive bands induced by sodium butyrate and the sustained Fos mRNA levels after induction suggested that the sodium butyrate-induced c-Fos protein could be non-functional in the autoregulation of the c-fos gene. Butyric Acid 70-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 2110635-1 1990 MK-801, a non-competitive N-methyl-D-aspartate (NMDA) receptor antagonist, leads to a dramatic induction of c-fos-like protein in neurons in deep layers of the neocortex, in dorsal and ventral midline thalamic nuclei and in neurons in the central grey of rat brain. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 2110635-2 1990 This induction of c-fos by MK-801 is dose-and time-dependent occurring within 2 h and dissipating by 24 h after injection (0.5-8.0 mg/kg, i.p.). Dizocilpine Maleate 27-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 2110635-3 1990 The mechanism of this paradoxical induction of c-fos by MK-801 is unclear; however, the pattern of induction appears to follow the distribution of the antagonist-preferring NMDA receptor site. Dizocilpine Maleate 56-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 2110636-0 1990 Opiates modify induction of c-fos proto-oncogene in the spinal cord of the rat following noxious stimulation. Opiate Alkaloids 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 2110636-1 1990 The expression of the proto-oncogene c-fos in neurons of the spinal cord dorsal horn of the rat following noxious thermal stimulation was compared in morphine- and ketamine-treated animals. Morphine 150-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-42 2110636-2 1990 Intravenous injection of morphine reduced the number of c-fos-positive neurons by up to 85% in laminae III-VI and X. Morphine 25-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 2110636-5 1990 The present data show that morphine suppresses the induction of c-fos. Morphine 27-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 1966043-1 1990 Deregulated c-fos expression in the rat pheochromocytoma cell line, PC-12, causes pronounced downregulation of nerve growth factor (NGF)-induced c-fos and c-jun activation, accompanied by a block in NGF-induced differentiation of PC-12 cells. PC 12 ester 68-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 2318256-3 1990 Administration of estradiol to ovariectomized rats determined a rapid and transient increase in the levels of c-fos mRNA in rat mid-brain and hippocampus, but not in the cerebellum (a brain area which does not express estrogen receptors). Estradiol 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 2318256-4 1990 Such a change in the levels of c-fos mRNA was not observed after administration of alpha-estradiol or other steroid hormones. alfatradiol 83-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 2318256-4 1990 Such a change in the levels of c-fos mRNA was not observed after administration of alpha-estradiol or other steroid hormones. Steroids 108-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 1690514-3 1990 Peak effects on c-fos mRNA occurred between 15 and 30 min and were completely gone after 2 h. The elevation in c-fos mRNA was, in part, dependent on protein kinase C, since phorbol myristate acetate (PMA) also elevated c-fos mRNA and further increased c-fos mRNA expression by endothelin, but the effects were not additive. Tetradecanoylphorbol Acetate 173-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 1690514-3 1990 Peak effects on c-fos mRNA occurred between 15 and 30 min and were completely gone after 2 h. The elevation in c-fos mRNA was, in part, dependent on protein kinase C, since phorbol myristate acetate (PMA) also elevated c-fos mRNA and further increased c-fos mRNA expression by endothelin, but the effects were not additive. Tetradecanoylphorbol Acetate 173-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 1690514-3 1990 Peak effects on c-fos mRNA occurred between 15 and 30 min and were completely gone after 2 h. The elevation in c-fos mRNA was, in part, dependent on protein kinase C, since phorbol myristate acetate (PMA) also elevated c-fos mRNA and further increased c-fos mRNA expression by endothelin, but the effects were not additive. Tetradecanoylphorbol Acetate 173-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 1690514-3 1990 Peak effects on c-fos mRNA occurred between 15 and 30 min and were completely gone after 2 h. The elevation in c-fos mRNA was, in part, dependent on protein kinase C, since phorbol myristate acetate (PMA) also elevated c-fos mRNA and further increased c-fos mRNA expression by endothelin, but the effects were not additive. Tetradecanoylphorbol Acetate 173-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 1690514-3 1990 Peak effects on c-fos mRNA occurred between 15 and 30 min and were completely gone after 2 h. The elevation in c-fos mRNA was, in part, dependent on protein kinase C, since phorbol myristate acetate (PMA) also elevated c-fos mRNA and further increased c-fos mRNA expression by endothelin, but the effects were not additive. Tetradecanoylphorbol Acetate 200-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 1690514-3 1990 Peak effects on c-fos mRNA occurred between 15 and 30 min and were completely gone after 2 h. The elevation in c-fos mRNA was, in part, dependent on protein kinase C, since phorbol myristate acetate (PMA) also elevated c-fos mRNA and further increased c-fos mRNA expression by endothelin, but the effects were not additive. Tetradecanoylphorbol Acetate 200-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 1690514-3 1990 Peak effects on c-fos mRNA occurred between 15 and 30 min and were completely gone after 2 h. The elevation in c-fos mRNA was, in part, dependent on protein kinase C, since phorbol myristate acetate (PMA) also elevated c-fos mRNA and further increased c-fos mRNA expression by endothelin, but the effects were not additive. Tetradecanoylphorbol Acetate 200-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 1966043-1 1990 Deregulated c-fos expression in the rat pheochromocytoma cell line, PC-12, causes pronounced downregulation of nerve growth factor (NGF)-induced c-fos and c-jun activation, accompanied by a block in NGF-induced differentiation of PC-12 cells. PC 12 ester 230-235 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 1966043-3 1990 Pronounced downregulation of c-fos, c-jun, and--to a lesser extent--jun-B was observed on treatment with NGF, bFGF, db cAMP, and Ca-ionophore, whereas EGF-induced activation of these early response genes was not inhibited in FOS-expressing PC-12 cells. Cyclic AMP 119-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 1966043-4 1990 Ca-ionophore- and db cAMP-induced egr-1 activation in PC-12 fos cells was completely inhibited. Cyclic AMP 21-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 1690127-4 1990 cAMP-mediated induction of the PDGF receptor results in enhanced, ligand dependent receptor autophosphorylation, and in enhanced PDGF activation of c-fos gene expression. Cyclic AMP 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 2109845-2 1990 Subcutaneous injection of 50 mg/kg capsaicin potently induced c-fos in the spinal cord on the first postnatal day (P1), whilst plantar injection of dilute formalin was much less effective until P3. Capsaicin 35-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 2105950-5 1990 In the presence of cycloheximide, a greater increase in c-fos mRNA was seen by stretching. Cycloheximide 19-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 1690514-3 1990 Peak effects on c-fos mRNA occurred between 15 and 30 min and were completely gone after 2 h. The elevation in c-fos mRNA was, in part, dependent on protein kinase C, since phorbol myristate acetate (PMA) also elevated c-fos mRNA and further increased c-fos mRNA expression by endothelin, but the effects were not additive. Tetradecanoylphorbol Acetate 200-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 2106434-0 1990 Luteinizing hormone-releasing hormone neurons express c-fos antigen after steroid activation. Steroids 74-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 2106434-6 1990 These data demonstrate that gonadal steroids, administered in a paradigm that predictably produces timed stimulation of LH release, induce c-fos in LHRH neurons. Steroids 36-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 2156188-0 1990 MK-801, an antagonist of NMDA receptors, inhibits injury-induced c-fos protein accumulation in rat brain. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 2156188-2 1990 C-fos protein was detected immunocytochemically using two different antibodies in formalin-fixed brain sections. Formaldehyde 82-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 2156188-3 1990 The N-methyl-D-aspartate (NMDA) antagonist MK-801 produced a dose- and time-dependent inhibition of c-fos protein accumulation in dentate granule cells and in neurons in the piriform cortex, but did not affect glial or ependymal c-fos protein accumulation. N-Methylaspartate 4-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 2156188-3 1990 The N-methyl-D-aspartate (NMDA) antagonist MK-801 produced a dose- and time-dependent inhibition of c-fos protein accumulation in dentate granule cells and in neurons in the piriform cortex, but did not affect glial or ependymal c-fos protein accumulation. N-Methylaspartate 4-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 229-234 2156188-3 1990 The N-methyl-D-aspartate (NMDA) antagonist MK-801 produced a dose- and time-dependent inhibition of c-fos protein accumulation in dentate granule cells and in neurons in the piriform cortex, but did not affect glial or ependymal c-fos protein accumulation. N-Methylaspartate 26-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 2156188-3 1990 The N-methyl-D-aspartate (NMDA) antagonist MK-801 produced a dose- and time-dependent inhibition of c-fos protein accumulation in dentate granule cells and in neurons in the piriform cortex, but did not affect glial or ependymal c-fos protein accumulation. N-Methylaspartate 26-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 229-234 2156188-3 1990 The N-methyl-D-aspartate (NMDA) antagonist MK-801 produced a dose- and time-dependent inhibition of c-fos protein accumulation in dentate granule cells and in neurons in the piriform cortex, but did not affect glial or ependymal c-fos protein accumulation. Dizocilpine Maleate 43-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 2156188-3 1990 The N-methyl-D-aspartate (NMDA) antagonist MK-801 produced a dose- and time-dependent inhibition of c-fos protein accumulation in dentate granule cells and in neurons in the piriform cortex, but did not affect glial or ependymal c-fos protein accumulation. Dizocilpine Maleate 43-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 229-234 2156188-4 1990 MK-801 at 4 mg/kg injected two hours before lesion inhibited c-fos accumulation. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 1966043-1 1990 Deregulated c-fos expression in the rat pheochromocytoma cell line, PC-12, causes pronounced downregulation of nerve growth factor (NGF)-induced c-fos and c-jun activation, accompanied by a block in NGF-induced differentiation of PC-12 cells. PC 12 ester 68-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-150 2108404-5 1990 Nonetheless, upon activating the pp60v-src protein kinase there is a marked and rapid increase in the ability of ts LA 29 Rat-1 nuclear extracts to retard the gel migration of oligonucleotides containing the AP-1 binding site, indicating that pp60v-src activity leads to an enhanced functioning of Fos and Jun related proteins that may, in turn, affect their transcriptional activation. Oligonucleotides 176-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 298-301 2105964-6 1990 c-fos transfected cells also had a faster growth rate than did control cells in serum-free medium supplemented with calcium chloride, lithium chloride, sodium selenite, hydrocortisone, and insulin. Calcium Chloride 116-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 2105964-6 1990 c-fos transfected cells also had a faster growth rate than did control cells in serum-free medium supplemented with calcium chloride, lithium chloride, sodium selenite, hydrocortisone, and insulin. Lithium Chloride 134-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 2105964-6 1990 c-fos transfected cells also had a faster growth rate than did control cells in serum-free medium supplemented with calcium chloride, lithium chloride, sodium selenite, hydrocortisone, and insulin. Sodium Selenite 152-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 2105964-6 1990 c-fos transfected cells also had a faster growth rate than did control cells in serum-free medium supplemented with calcium chloride, lithium chloride, sodium selenite, hydrocortisone, and insulin. Hydrocortisone 169-183 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 2404011-1 1990 12-O-Tetradecanoylphorbol-13-acetate (TPA) activated the c-fos gene enhancer linked to the chloramphenicol acetyltransferase or luciferase reporter gene in the wild type PC-12 cells but not in the variant PC-12 cells that originated from the wild type cells. Tetradecanoylphorbol Acetate 0-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 2404011-1 1990 12-O-Tetradecanoylphorbol-13-acetate (TPA) activated the c-fos gene enhancer linked to the chloramphenicol acetyltransferase or luciferase reporter gene in the wild type PC-12 cells but not in the variant PC-12 cells that originated from the wild type cells. Tetradecanoylphorbol Acetate 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 2404011-2 1990 Transfection of the c-Ha-rasval12 complementary DNA (cDNA) or addition of dibutyryl cAMP to the wild type PC-12 cells as well as to the variant PC-12 cells activated the c-fos gene enhancer. Cyclic AMP 84-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-175 2404011-3 1990 Prolonged treatment of the wild type PC-12 cells with phorbol-12,13-dibutyrate caused down-regulation of protein kinase C. In these cells, TPA did not stimulate the c-fos gene enhancer any more, but transfection of the c-Ha-rasval12 cDNA still stimulated the c-fos gene enhancer to the same extent as induced in the control cells. Phorbol 12,13-Dibutyrate 54-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 259-264 2127224-6 1990 A distinct increase in the expression of the c-fos gene in the brain cortex was observed in offspring of chronically alcohol treated rats after administration of ethanol. Alcohols 117-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 2105102-3 1990 While Ki-ras mRNA levels remained relatively unchanged, mRNAs for fos and sis increased significantly during the course of butyrate induced differentiation. Butyrates 123-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 2105102-4 1990 c-fos induction can be detected 30 min after butyrate addition, a peak level (greater than 20 fold) was reached at 2 h, with a subsequent gradual decline. Butyrates 45-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 2105726-4 1990 Expression of the c-fos gene may be functionally linked to the nocturnal changes in pineal indole metabolism. indole 91-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 2105102-0 1990 Induction of fos and sis proto-oncogenes and genes of the extracellular matrix proteins during butyrate induced glioma differentiation. Butyrates 95-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 2127224-6 1990 A distinct increase in the expression of the c-fos gene in the brain cortex was observed in offspring of chronically alcohol treated rats after administration of ethanol. Ethanol 162-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 2127224-7 1990 The impairments in regulating c-fos gene activity caused by ethanol administration suggest that chronic alcohol treatment of animals may influence the functions of the genome of offspring not subjected to chronic alcohol treatment. Ethanol 60-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 2127224-7 1990 The impairments in regulating c-fos gene activity caused by ethanol administration suggest that chronic alcohol treatment of animals may influence the functions of the genome of offspring not subjected to chronic alcohol treatment. Alcohols 104-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 2127224-7 1990 The impairments in regulating c-fos gene activity caused by ethanol administration suggest that chronic alcohol treatment of animals may influence the functions of the genome of offspring not subjected to chronic alcohol treatment. Alcohols 213-220 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 2129102-2 1990 Addition of fetal calf serum (FCS) to serum-starved cells in the presence of cycloheximid induced a modest increase in c-fos and c-jun mRNA levels, whereas growth hormone (GH) in the presence of cycloheximid had little or no effect, when added to RIN 5AH cells maintained in 0.5% FCS for 2 days prior to stimulation. Cycloheximide 77-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 2129102-3 1990 Activation of protein kinase C by phorbol ester lead to increased c-jun and c-fos mRNA levels, whereas activation of adenylate cyclase by forskolin increased c-fos mRNA levels. Phorbol Esters 34-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 2129102-3 1990 Activation of protein kinase C by phorbol ester lead to increased c-jun and c-fos mRNA levels, whereas activation of adenylate cyclase by forskolin increased c-fos mRNA levels. Colforsin 138-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 2279259-3 1990 FRTL5 cells were stably transfected with plasmid constructs that transcribe, under the control of the dexamethasone-inducible MMTV promoter, a 5"-fragment (84 nucleotides) of c-fos mRNA, either in the sense or in the antisense orientation. Dexamethasone 102-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 175-180 2104799-3 1990 This c-fos expression can be specifically suppressed by coinjection of a double-stranded oligonucleotide which corresponds to the serum response element (SRE) present in the c-fos promoter, implying that ras utilizes a pathway which activates the binding of serum response factor(s) (SRF) to SRE to induce c-fos transcription. Oligonucleotides 89-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 5-10 2104799-3 1990 This c-fos expression can be specifically suppressed by coinjection of a double-stranded oligonucleotide which corresponds to the serum response element (SRE) present in the c-fos promoter, implying that ras utilizes a pathway which activates the binding of serum response factor(s) (SRF) to SRE to induce c-fos transcription. Oligonucleotides 89-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 174-179 2104799-3 1990 This c-fos expression can be specifically suppressed by coinjection of a double-stranded oligonucleotide which corresponds to the serum response element (SRE) present in the c-fos promoter, implying that ras utilizes a pathway which activates the binding of serum response factor(s) (SRF) to SRE to induce c-fos transcription. Oligonucleotides 89-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 174-179 2279259-9 1990 Taken together, our results provide circumstantial evidence that c-fos, at least in part, may play a role in TSH-mediated thyroid cell growth. Thyrotropin 109-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 1966822-1 1990 Rats injected with haloperidol, which binds to both D2 dopamine and sigma receptors or the specific D2 dopamine receptor antagonist YM 09151-2, but not the specific D1 dopamine receptor antagonist SCH 23390, showed induction of c-fos protein and c-fos-related antigens in striatal neurons. Haloperidol 19-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 228-233 1688935-0 1990 Systemic morphine suppresses noxious stimulus-evoked Fos protein-like immunoreactivity in the rat spinal cord. Morphine 9-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 1688935-3 1990 In this study, we examined the effect of systemic morphine on Fos-like immunoreactivity (FLI) evoked in the formalin test, a widely used model of persistent pain. Morphine 50-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 1688935-3 1990 In this study, we examined the effect of systemic morphine on Fos-like immunoreactivity (FLI) evoked in the formalin test, a widely used model of persistent pain. Formaldehyde 108-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 2163480-0 1990 The fos proto-oncogene protein:regulation by morphine in the rat hypothalamus. Morphine 45-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-7 2163480-2 1990 We have previously demonstrated that morphine rapidly and transiently increases expression of the proto-oncogene c-fos in the rat caudate-putamen. Morphine 37-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-118 2163480-3 1990 This regulation of the c-fos gene by morphine may represent a portion of the intracellular cascade coupling activation of opiate receptors on the cell surface to subsequent alterations in neuropeptide gene expression. Morphine 37-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 2163480-4 1990 In the present study, we have focussed on effects of morphine on c-fos expression in the ventromedial hypothalamus, which contains estrogen-concentrating neurons and a large number of neurons expressing the opioid proenkephalin and Proopiomelanocortin. Morphine 53-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 2215924-3 1990 Subcutaneous injection of dilute formalin produced a similar pattern of immunostaining at 2 h, with a greater proportion of Fos-positive neurons in laminae III-VIII than with heat. Formaldehyde 33-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-127 2215924-4 1990 With a survival time of 8 h following formalin injection, Fos immunoreactivity was virtually absent from the spinal cord. Formaldehyde 38-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 2215924-7 1990 The number of Fos-positive cells seen at 8 h was increased by local anaesthetic blockade of the peripheral nerve after stimulation, and reduced by continuous barbiturate anaesthesia. barbituric acid 158-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 1966822-1 1990 Rats injected with haloperidol, which binds to both D2 dopamine and sigma receptors or the specific D2 dopamine receptor antagonist YM 09151-2, but not the specific D1 dopamine receptor antagonist SCH 23390, showed induction of c-fos protein and c-fos-related antigens in striatal neurons. Haloperidol 19-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 246-251 1966822-4 1990 In time-course immunocytochemical studies, the c-fos protein was induced maximally by 1 h and had returned to baseline by 24 h. However, c-fos protein-related antigens were induced maximally after 2 h and remained elevated for at least three days after haloperidol injection. Haloperidol 253-264 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 1966822-4 1990 In time-course immunocytochemical studies, the c-fos protein was induced maximally by 1 h and had returned to baseline by 24 h. However, c-fos protein-related antigens were induced maximally after 2 h and remained elevated for at least three days after haloperidol injection. Haloperidol 253-264 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 1966822-5 1990 Furthermore, the c-fos protein-specific antiserum detected two to three times fewer immunopositive striatal cells than the antiserum which detected both c-fos protein-related antigens and c-fos protein in haloperidol-treated rats. Haloperidol 205-216 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 1966822-7 1990 In some striatal sections from haloperidol-injected rats immunostained with the antiserum which recognizes both c-fos protein and c-fos protein-related antigens, there were large areas of immunopositive neurons interspersed with "areas" of striatum devoid of immunostaining. Haloperidol 31-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 1966822-7 1990 In some striatal sections from haloperidol-injected rats immunostained with the antiserum which recognizes both c-fos protein and c-fos protein-related antigens, there were large areas of immunopositive neurons interspersed with "areas" of striatum devoid of immunostaining. Haloperidol 31-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 33773180-5 2021 The results showed that DEHP promoted rat pituitary tumor (GH3) cell proliferation and c-fos gene expression at environment level concentrations (2 and 5 mumol/L) in a manner similar to that of the natural thyroid hormone 3,3",5-triiodo-L-thyronine (T3). Diethylhexyl Phthalate 24-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 33773180-8 2021 Moreover, DEHP activated the downstream extracellular regulated protein kinase (ERK1/2) pathway, upregulating the gene expression of raf-1, MEK-1 and MAPK1 and increasing the protein levels of p-ERK; interestingly, ERK1/2 activation and c-fos upregulation induced by DEHP were attenuated by PD98059 (an ERK1/2 inhibitor). Diethylhexyl Phthalate 10-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 237-242 33773180-6 2021 The macromolecule DEHP-BSA cannot pass through the cell membrane to interact with nuclear receptors but upregulated the c-fos gene expression when administered at concentrations comparable to DEHP concentrations; molecular docking demonstrated that DEHP has affinity for the membrane receptor integrin alphavbeta3; DEHP at 2 mumol/L upregulated the beta3 gene expression in GH3 cells; after the addition of integrin alphavbeta3-inhibiting RGD peptide, DEHP-induced c-fos gene upregulation decreased. Diethylhexyl Phthalate 18-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 33773180-6 2021 The macromolecule DEHP-BSA cannot pass through the cell membrane to interact with nuclear receptors but upregulated the c-fos gene expression when administered at concentrations comparable to DEHP concentrations; molecular docking demonstrated that DEHP has affinity for the membrane receptor integrin alphavbeta3; DEHP at 2 mumol/L upregulated the beta3 gene expression in GH3 cells; after the addition of integrin alphavbeta3-inhibiting RGD peptide, DEHP-induced c-fos gene upregulation decreased. Diethylhexyl Phthalate 18-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 465-470 33806698-8 2021 Similarly, c-Fos, p-ERK, and p-NR2B levels were significantly higher in glutamate-treated PC12 neuronal cells but were not affected by Nfat5 silencing in glutamate-treated PC12 cells. Glutamic Acid 72-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 33811839-8 2021 The pain score and c-fos expression in the spinal cord were highest in the 0.9 mg capsaicin group and lowest in the normal saline and vehicle groups (P < 0.05). Capsaicin 82-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 33811839-9 2021 Intrathecal morphine significantly decreased the pain score (P < 0.05) and c-fos expression in the spinal cord (P < 0.05). Morphine 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 2261575-0 1990 Sodium butyrate inhibits c-myc and stimulates c-fos expression in all the steps of the cell-cycle in hepatoma tissue cultured cells. Butyric Acid 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 2261575-1 1990 Sodium butyrate decreases the c-myc mRNA and increases the c-fos transcript level in HTC cells. Butyric Acid 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 2261575-3 1990 Actinomycin D suppresses the effect on c-fos. Dactinomycin 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 2261575-5 1990 Sodium butyrate suppresses the effect on c-myc and potentializes the effect on c-fos mRNAs. Butyric Acid 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 25336083-9 2014 Subsequent post hoc test (Bonferroni"s) revealed that as compared to ACSF + W group, c-Fos expression was significantly increased in the shell of NAc of rats in all 3 (EtOH, NiC, and NiC + EtOH) groups with maximal increase observed in NiC + EtOH group. Ethanol 168-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 33799856-2 2021 These processes can be evaluated based on the decline in the frequency of spontaneous nystagmus (SN) and the number of MK801-induced Fos-positive neurons in the contralateral medial vestibular nucleus (contra-MVe) in rats. Dizocilpine Maleate 119-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-136 33799856-8 2021 The number of MK801-induced Fos-positive neurons in contra-MVe significantly decreased on days 7, 10, and 12 after UL in a dose-dependent manner in the betahistine-treated rats, more so than in the saline-treated rats. Dizocilpine Maleate 14-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 33799856-8 2021 The number of MK801-induced Fos-positive neurons in contra-MVe significantly decreased on days 7, 10, and 12 after UL in a dose-dependent manner in the betahistine-treated rats, more so than in the saline-treated rats. Betahistine 152-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 25336083-9 2014 Subsequent post hoc test (Bonferroni"s) revealed that as compared to ACSF + W group, c-Fos expression was significantly increased in the shell of NAc of rats in all 3 (EtOH, NiC, and NiC + EtOH) groups with maximal increase observed in NiC + EtOH group. Ethanol 189-193 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 25336083-9 2014 Subsequent post hoc test (Bonferroni"s) revealed that as compared to ACSF + W group, c-Fos expression was significantly increased in the shell of NAc of rats in all 3 (EtOH, NiC, and NiC + EtOH) groups with maximal increase observed in NiC + EtOH group. Ethanol 189-193 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 25336083-10 2014 CONCLUSIONS: The results suggest the following: (i) BF nicotine infusion induced c-Fos in both core and the shell region of NAc at levels comparable to those observed after systemic alcohol administration; (ii) BF nicotine infusion with systemic alcohol induced a significant additive increase in c-Fos expression only in the NAc shell region. Nicotine 55-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 25336083-10 2014 CONCLUSIONS: The results suggest the following: (i) BF nicotine infusion induced c-Fos in both core and the shell region of NAc at levels comparable to those observed after systemic alcohol administration; (ii) BF nicotine infusion with systemic alcohol induced a significant additive increase in c-Fos expression only in the NAc shell region. Nicotine 55-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 297-302 14556240-7 2003 We also show that subchronic fluoxetine and olanzapine exposure suppresses the induction of two immediate-early gene transcription factors (e.g., pCREB and FOS) that are associated with long-lasting changes in synaptic efficacy and structural modifications in the prefrontal cortex, piriform cortex, and hippocampus. Fluoxetine 29-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-159 34619303-12 2022 Besides, a negative correlation between the fear response and c-Fos expression in the dHC CA3/CA1 and vHC CA1/DG was observed after chronic FLX treatment. Fluoxetine 140-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 34813109-11 2022 As expected, hypoxia (8% O2 - 3 hours) induced a robust increase in fos expression within the catecholaminergic C1 and A5 regions of the brainstem. Oxygen 25-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 24631673-9 2014 Acute stimulation of SNc by picrotoxin increased cFOS-IR in the vermis and cerebellar hemispheres. Picrotoxin 28-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-53 24631673-11 2014 After 14days of haloperidol treatment, the vermis and cerebellar hemispheres showed an opposite regulation of cFOS expression. Haloperidol 16-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-114 16442086-11 2006 The difference in the distribution pattern of pERK- and Fos protein-LI cells in the Vi/Vc zone suggests their differential temporal expression profiles after capsaicin. Capsaicin 158-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 34942196-8 2022 In addition, the number of c-Fos and POMC colocalizations in the HTa group was higher than that in the CTa group. hta 65-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 34942196-8 2022 In addition, the number of c-Fos and POMC colocalizations in the HTa group was higher than that in the CTa group. Chlorotrianisene 103-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 34737150-8 2022 At the molecular level, perindopril downregulated the TLR4/NF-kappaB and c-Fos/c-Jun pathways in inflamed intestine of rats. Perindopril 24-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 34737150-11 2022 CONCLUSIONS: Perindopril demonstrated notable mitigation of MTX-induced intestinal injury through suppression of TLR4/NF-kappaB and c-Fos/c-Jun pathways alongside the augmentation of PPAR-gamma/SIRT1 cytoprotective signals. Perindopril 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 14556240-7 2003 We also show that subchronic fluoxetine and olanzapine exposure suppresses the induction of two immediate-early gene transcription factors (e.g., pCREB and FOS) that are associated with long-lasting changes in synaptic efficacy and structural modifications in the prefrontal cortex, piriform cortex, and hippocampus. Olanzapine 44-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-159 34813637-5 2021 In comparison with the sham group, propionate significantly increased the parasympathetic components heart rate variability (HRV) and acetylcholine levels, prolonged cardiac repolarization, induced STAT3 phosphorylation and up-regulated the c-fos expression in nodose ganglia and solitary nucleus. Propionates 35-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 241-246 34624416-0 2021 Exposure to Di-(2-ethylhexyl) phthalate reduces secretion of GDNF via interfering with estrogen pathway and downregulating ERK/c-fos signaling pathway in astrocytes. Diethylhexyl Phthalate 12-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 34779255-7 2022 CCK reduced the number of c-Fos-positive cells in the median preoptic area (by ~70%) but increased it in the dorsal hypothalamic area and in the rostral raphe pallidus (by ~50% in both); all these changes were all completely blocked with metamizol. Dipyrone 238-247 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 34942451-11 2021 Interestingly, lower levels of GFAP, c-Fos and 3-NT were observed in animals receiving beta-caryophyllene and pregabalin treatments. caryophyllene 87-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 34942451-11 2021 Interestingly, lower levels of GFAP, c-Fos and 3-NT were observed in animals receiving beta-caryophyllene and pregabalin treatments. Pregabalin 110-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 34624416-5 2021 Our results showed that DEHP exposure reduced GDNF levels and downregulated the ERK/c-fos signaling pathway in the cerebral cortex of male, but not female, offspring. Diethylhexyl Phthalate 24-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 34624416-7 2021 MEHP also decreased p300 levels and downregulated the ERK/c-fos signaling pathway in primary astrocytes. mono-(2-ethylhexyl)phthalate 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 34624416-8 2021 Honokiol restored GDNF levels following MEHP exposure by activating the ERK/c-fos signaling pathway, while the inhibitor U0126 further reduced the GDNF levels. honokiol 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 34624416-8 2021 Honokiol restored GDNF levels following MEHP exposure by activating the ERK/c-fos signaling pathway, while the inhibitor U0126 further reduced the GDNF levels. mono-(2-ethylhexyl)phthalate 40-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 34624416-9 2021 These results suggested that DEHP exposure could interfere with the normal effects of estrogen in the brain and downregulate the ERK/c-fos signaling pathway to decrease the GDNF secretion from astrocytes in the cerebral cortex. Diethylhexyl Phthalate 29-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 34182089-9 2021 This resulted in a significant decrease of photo-stimulated neurons (immunoreactivity to c-Fos) in the SCN of the metal-exposed animals. Metals 114-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 34873411-5 2021 When the LIFU intensity was more than 0.5 MPa, LIFU stimulation induced soleus muscle contraction and increased the EMG amplitudes (P < 0.05) and the number of c-fos- and GAD65-positive cells (P < 0.05). lifu 9-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 34873411-5 2021 When the LIFU intensity was more than 0.5 MPa, LIFU stimulation induced soleus muscle contraction and increased the EMG amplitudes (P < 0.05) and the number of c-fos- and GAD65-positive cells (P < 0.05). lifu 47-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 34869766-0 2021 Effects of Ozone on Hippocampus BDNF and Fos Expressions in Rats with Chronic Compression of Dorsal Root Ganglia. Ozone 11-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 34869766-1 2021 The effects of ozone on hippocampal expression levels of brain-derived neurotrophic factor (BDNF) and c-fos protein (Fos) were evaluated in rats with chronic compression of dorsal root ganglia (CCD). Ozone 15-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-115 34869766-1 2021 The effects of ozone on hippocampal expression levels of brain-derived neurotrophic factor (BDNF) and c-fos protein (Fos) were evaluated in rats with chronic compression of dorsal root ganglia (CCD). Ozone 15-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 34869766-7 2021 The TWLs and MWTs of CCD model rats that received ozone were lower with decreased BDNF and increased Fos expression levels, on day 21 after the operation, compared to those of the sham group (P < 0.05). Ozone 50-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-104 34869766-8 2021 The TWLs and MWTs of the CCD + AO3 and CCD + BO3 groups were higher with increased BDNF and decreased Fos expression levels, on day 21 after the operation, compared to those of the CCD group (P < 0.05). boric acid 45-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 34869766-9 2021 The TWLs were longer and the MWTs were higher in the CCD + BO3 group at each time point with increased BDNF and decreased Fos expression levels, on day 21 after the operation, compared to those of the CCD + AO3 group (P < 0.05). boric acid 59-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 34869766-11 2021 The mechanism of action for ozone is likely associated with changes in BDNF and Fos expression levels in the hippocampus. Ozone 28-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 34772825-7 2021 Sinomenine treatment reduced RIP3, p-JNK and c-Fos levels in the spinal cords of CCI rats. sinomenine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 34431619-7 2021 The results indicated that treatment with EA and fluoxetine can reverse the CUMS-induced depression-like behaviors in rats and can up-regulate the expression levels of c-Jun in the hypothalamus, c-Fos in the pituitary gland, and c-Fos and AP-1 in the serum. Fluoxetine 49-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-200 34431619-7 2021 The results indicated that treatment with EA and fluoxetine can reverse the CUMS-induced depression-like behaviors in rats and can up-regulate the expression levels of c-Jun in the hypothalamus, c-Fos in the pituitary gland, and c-Fos and AP-1 in the serum. Fluoxetine 49-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 229-234 34665381-9 2021 In the QNP-Paired group, exposure to the young male-paired odor evoked more Fos-IR in limbic, hypothalamic and cortical areas, but no differences in serum testosterone were observed. (2S)-1-(nonanoyloxy)-3-(phosphonooxy)propan-2-yl tetradecanoate 7-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 34730349-9 2021 Our results demonstrate (i) the presence of proton pumps in the LC neurons, (ii) that the microinjection of lansoprazole into the LC reduced the number of cFOS+ve-TH+ve double-labeled neurons in the LC by 52.6% (p < 0.001) compared to the vehicle and (iii) that low and high doses of lansoprazole significantly increased REM sleep by 32% (p < 0.001) and 60% (p < 0.001), respectively, compared to the vehicle. Lansoprazole 108-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-159 34730349-9 2021 Our results demonstrate (i) the presence of proton pumps in the LC neurons, (ii) that the microinjection of lansoprazole into the LC reduced the number of cFOS+ve-TH+ve double-labeled neurons in the LC by 52.6% (p < 0.001) compared to the vehicle and (iii) that low and high doses of lansoprazole significantly increased REM sleep by 32% (p < 0.001) and 60% (p < 0.001), respectively, compared to the vehicle. Lansoprazole 284-296 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-159 34768597-8 2021 The 4V OT also induced Fos in tyrosine hydroxylase (TH; marker of catecholamine neurons) (+) neurons (25 +- 7%) relative to vehicle (0.8 +- 0.3%). Catecholamines 66-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 34347648-12 2021 bupivacaine similarly inhibited c-Fos activation in NTS. Bupivacaine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 34347648-21 2021 Our study underlines the role of the vagus nerve in the transmission of an acute visceral pain message and confirmed that systemic bupivacaine prevents noxious stimuli by inhibiting c-Fos and microglia activation. Bupivacaine 131-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 182-187 34347648-23 2021 bupivacaine similarly inhibited c-Fos and microglial activation both in cord and in the NTS. Bupivacaine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 33818520-0 2021 Magnesium acetyltaurate prevents retinal damage and visual impairment in rats through suppression of NMDA-induced upregulation of NF-kappaB, p53 and AP-1 (c-Jun/c-Fos). acetyltaurine 0-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 33818520-0 2021 Magnesium acetyltaurate prevents retinal damage and visual impairment in rats through suppression of NMDA-induced upregulation of NF-kappaB, p53 and AP-1 (c-Jun/c-Fos). N-Methylaspartate 101-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 33818520-2 2021 The current study investigated the involvement of nuclear factor kappa-B (NF-kappaB), p53 and AP-1 family members (c-Jun/c-Fos) in neuroprotection by MgAT against NMDA-induced retinal damage. acetyltaurine 150-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-126 33818520-10 2021 MgAT abolished NMDA-induced increase of retinal phospho-NF-kappaB, phospho-p53 and AP-1 expression and suppressed NMDA-induced transcriptional activity of NF-kappaB, p53 and AP-1 family members (c-Jun/c-Fos). N-Methylaspartate 15-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 201-206 33818520-10 2021 MgAT abolished NMDA-induced increase of retinal phospho-NF-kappaB, phospho-p53 and AP-1 expression and suppressed NMDA-induced transcriptional activity of NF-kappaB, p53 and AP-1 family members (c-Jun/c-Fos). N-Methylaspartate 114-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 201-206 33818520-12 2021 In conclusion, pre-treatment with MgAT prevents NMDA induced retinal injury by inhibiting NMDA-induced neuronal apoptosis via downregulation of transcriptional activity of NF-kappaB, p53 and AP-1-mediated c-Jun/c-Fos. N-Methylaspartate 90-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 211-216 34639188-5 2021 Moreover, we analysed by immunohistochemistry the expression of the immediate-early gene c-Fos (c-Fos IR) after the administration of GAL(1-15)+ESC in OBX rats in several nuclei involved in MDD. cyclohexenoesculetin-beta-galactoside 134-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 34744653-16 2021 The results from the Fos data suggest that higher alcohol seeking in approach-avoidance motivational conflict is associated with activation of sub-cortical regions but not cortical regions. Alcohols 50-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 34744735-7 2021 In addition, emodin can significantly reduce the number of c-Fos positive cells and the IOD value. Emodin 13-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 34755664-8 2021 In the rat models of COPD, treatment with Liuwei Buqi capsule significantly improved pulmonary function, alleviated lung pathologies, reduced serum levels of IL-1beta, TNF-alpha, and NF-kappaB (P < 0.05) and pulmonary expressions of JNK, c-JUN, and c-FOS (P < 0.01) protein, and increased pulmonary expression of IkappaBalpha (P < 0.01). liuwei buqi capsule 42-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 249-254 34218375-9 2021 Furthermore, deltamethrin increased mRNA expression levels of PARP-1, VEGF, and immunohistochemical expressions of c-fos in the tissues. decamethrin 13-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 34406994-6 2021 RESULTS: The c-Fos and NF-kappaB gene expression levels in protective group are lower than those in the irradiated group after MgSO4 treatment. Magnesium Sulfate 127-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 34406994-10 2021 The protective mechanisms of Mg2+ in the hippocampi from a variety of brain activity indicators including the c-Fos and NF-kappaB genes. magnesium ion 29-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 34639188-5 2021 Moreover, we analysed by immunohistochemistry the expression of the immediate-early gene c-Fos (c-Fos IR) after the administration of GAL(1-15)+ESC in OBX rats in several nuclei involved in MDD. cyclohexenoesculetin-beta-galactoside 134-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 34243110-6 2021 Histological analyses revealed that NMU increased Fos-IR in the CA1 region, but reduced the proportion of Fos-IR colocalized with glutamic acid decarboxylase (a GABA neuron marker). Glutamic Acid 130-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 34243110-6 2021 Histological analyses revealed that NMU increased Fos-IR in the CA1 region, but reduced the proportion of Fos-IR colocalized with glutamic acid decarboxylase (a GABA neuron marker). gamma-Aminobutyric Acid 161-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 34311052-8 2021 Central dopamine depletion induced an increase in excitation as reveled by an increase in cFOS expression upon acetone stimulus and the presence of cold allodynia. Dopamine 8-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-94 34242672-0 2021 Ventral tegmental area serotonin 5-HT1A receptors and corticolimbic cFos/BDNF/GFAP signaling pathways mediate dextromethorphan/morphine anti-allodynia. Dextromethorphan 110-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-72 34242672-0 2021 Ventral tegmental area serotonin 5-HT1A receptors and corticolimbic cFos/BDNF/GFAP signaling pathways mediate dextromethorphan/morphine anti-allodynia. Morphine 127-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-72 34242672-7 2021 Dextromethorphan/morphine-induced anti-allodynia was accompanied by the decrease of hippocampus/amygdala/PFC GFAP and amygdala cFos expressions. Dextromethorphan 0-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-131 34242672-7 2021 Dextromethorphan/morphine-induced anti-allodynia was accompanied by the decrease of hippocampus/amygdala/PFC GFAP and amygdala cFos expressions. Morphine 17-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-131 34311052-8 2021 Central dopamine depletion induced an increase in excitation as reveled by an increase in cFOS expression upon acetone stimulus and the presence of cold allodynia. Acetone 111-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-94 34332026-5 2021 We also examined the change of c-Fos expression induced by isoflurane exposure in cerebellum granule cells of both mutant and WT rats. Isoflurane 59-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 34332026-7 2021 Moreover, isoflurane exposure induced a greater reduction in c-Fos expression in cerebellum granule cells of Gabra6100Q rats than WT rats. Isoflurane 10-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 34523299-13 2021 The CMS decreased the c-Fos immunoreactivity in the PFC in both HAL- and ARI-treated animals. Haloperidol 64-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 34523299-13 2021 The CMS decreased the c-Fos immunoreactivity in the PFC in both HAL- and ARI-treated animals. Aripiprazole 73-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 34523299-14 2021 In the HIP, HAL increased the c-Fos immunoreactivity that was again diminished in animals exposed to CMS. Haloperidol 12-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 34523299-14 2021 In the HIP, HAL increased the c-Fos immunoreactivity that was again diminished in animals exposed to CMS. N-Carbamoylsarcosine 101-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 34224765-9 2021 Formalin increased TREK-1 expression at 1 and 6 days in DRG and dorsal spinal cord in rats, whereas that it increased c-fos expression at the DRG. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 34224765-10 2021 Intrathecal repeated transfection of rats with S300A and S333A or injection with BL-1249 reduced formalin-induced enhanced c-fos expression. Formaldehyde 97-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 34326141-6 2021 The results showed that intraperitoneal injection of acetaldehyde increased cFos protein expression within the LHb and that intra-LHb infusion of acetaldehyde induced conditioned place aversion in male rats. Acetaldehyde 53-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-80 34490827-0 2021 Sucrose drinking mimics effects of nucleus accumbens micro-opioid receptor stimulation on fat intake and brain c-Fos-expression. Sucrose 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 34539968-15 2021 Puerarin was the active ingredient derived from Puerariae Radix which exhibited the antidepression effect by improving the viability of cell, reducing cell apoptosis, regulating ROS production, increasing protein expressions of AKT1 and FOS, and reducing protein expressions of CASP3, STAT3, and TNF-alpha. puerarin 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 237-240 34552469-9 2021 Analysis of c-Fos expression following systemic injections of angiotensin II or hypertonic NaCl reveals distinct neuronal populations responding to these stimuli. Sodium Chloride 91-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 34491515-14 2022 In addition, DEX reduced the number of c-Fos positive cells in dentate gyrus and the hippocampal CA1 and CA3 regions. Dexmedetomidine 13-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 34490827-7 2021 Both intra-NAC DAMGO infusion and sucrose consumption in the absence of DAMGO infusion had no effect on c-Fos-expression in orexin neurons and the central amygdala but increased c-Fos-expression in the NAC as well as the basolateral amygdala.Discussion: In conclusion, we confirm that sucrose drinking stimulates fat intake, likely through the release of endogenous opioids. Sucrose 34-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 178-183 34490827-7 2021 Both intra-NAC DAMGO infusion and sucrose consumption in the absence of DAMGO infusion had no effect on c-Fos-expression in orexin neurons and the central amygdala but increased c-Fos-expression in the NAC as well as the basolateral amygdala.Discussion: In conclusion, we confirm that sucrose drinking stimulates fat intake, likely through the release of endogenous opioids. Sucrose 285-292 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 178-183 34403389-4 2021 METHODS: The c-Fos staining was used to observe the activity changes of orexinergic neurons in the PeFLH and their efferent projection regions under desflurane anesthesia. Desflurane 149-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 34403389-7 2021 RESULTS: Desflurane anesthesia inhibited the activity of orexinergic neurons in the PeFLH, as well as the neuronal activity in PVT, basal forebrain, dorsal raphe nucleus, and ventral tegmental area, as demonstrated by c-Fos staining. Desflurane 9-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 218-223 34197938-8 2021 Analysis of immediate early gene expression revealed that morphine reduced the expression of cfos in the prefrontal cortex of both saline- and VPA-exposed rats and reduced expression of cfos and junb in the hippocampus of VPA-exposed rats only. Valproic Acid 222-225 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 186-190 34081989-9 2021 On the other hand, TBARS and TNF-alpha levels were reduced and NF-kB/p65 and TLR-4 expressions were decreased together with increase in total -SH levels among CLP + FOS (500 mg/kg i.p.) Thiobarbituric Acid Reactive Substances 19-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-168 34197938-8 2021 Analysis of immediate early gene expression revealed that morphine reduced the expression of cfos in the prefrontal cortex of both saline- and VPA-exposed rats and reduced expression of cfos and junb in the hippocampus of VPA-exposed rats only. Morphine 58-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-97 34197938-8 2021 Analysis of immediate early gene expression revealed that morphine reduced the expression of cfos in the prefrontal cortex of both saline- and VPA-exposed rats and reduced expression of cfos and junb in the hippocampus of VPA-exposed rats only. Morphine 58-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 186-190 34197938-8 2021 Analysis of immediate early gene expression revealed that morphine reduced the expression of cfos in the prefrontal cortex of both saline- and VPA-exposed rats and reduced expression of cfos and junb in the hippocampus of VPA-exposed rats only. Valproic Acid 143-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-97 34161572-4 2021 Administration of 2-deoxy-D-glucose (2DG), an inhibitor of glucose utilization, into the fourth ventricle (4V) of male rats for 0.5 h induced mRNA expression of c-fos, a marker for cellular activation, in ependymal cells in the 4V, but not in the lateral ventricle, the third ventricle or the central canal without a significant change in blood glucose and testosterone levels. Glucose 28-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 34161572-4 2021 Administration of 2-deoxy-D-glucose (2DG), an inhibitor of glucose utilization, into the fourth ventricle (4V) of male rats for 0.5 h induced mRNA expression of c-fos, a marker for cellular activation, in ependymal cells in the 4V, but not in the lateral ventricle, the third ventricle or the central canal without a significant change in blood glucose and testosterone levels. Deoxyglucose 37-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 34161572-4 2021 Administration of 2-deoxy-D-glucose (2DG), an inhibitor of glucose utilization, into the fourth ventricle (4V) of male rats for 0.5 h induced mRNA expression of c-fos, a marker for cellular activation, in ependymal cells in the 4V, but not in the lateral ventricle, the third ventricle or the central canal without a significant change in blood glucose and testosterone levels. Glucose 59-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 34522608-4 2021 We used an ex vivo imaging method in the Wistar Fos-LacZ transgenic rat, to detect neuronal ensembles in medial prefrontal cortex (mPFC) following cocaine administration or a shock-tone paired stimulus. Cocaine 147-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 34532455-12 2021 Ligustrazine could inhibit over-expression of P2X3, TRPV1, c-fos, and ERK in the TG nerve of NTG-induced migraine rats. tetramethylpyrazine 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 34532455-13 2021 Conclusions: Our results demonstrated that ligustrazine had potent activity against NTG-induced migraine rats through inhibition of the c-fos/ERK signaling pathway. tetramethylpyrazine 43-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 34253253-0 2021 Assessing the treatment of cannabidiolic acid methyl ester: a stable synthetic analogue of cannabidiolic acid on c-Fos and NeuN expression in the hypothalamus of rats. CBDA-ME 27-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 34373508-6 2021 We observed that microinjection of the selective CB1 receptor antagonist AM251 into the BNST decreased the number of Fos-immunoreactive cells within the LH of rats submitted to acute restraint stress. AM 251 73-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 34286313-7 2021 In the mPFC, 10 mg/kg ketamine reduced pERK levels in male rats while 40 mg/kg ketamine increased c-Fos levels in male and female rats. Ketamine 22-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 34286313-7 2021 In the mPFC, 10 mg/kg ketamine reduced pERK levels in male rats while 40 mg/kg ketamine increased c-Fos levels in male and female rats. Ketamine 79-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 34286313-8 2021 Female rats in proestrus/estrus phases showed greater ketamine-induced c-Fos elevation as compared to those in diestrus phase. Ketamine 54-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 34286313-9 2021 In the amygdala, 10 and 40 mg/kg ketamine increased c-Fos levels in female, but not male, rats. Ketamine 33-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 34402265-5 2021 Compared with surgery group, propofol group and surgery with propofol group showed increased TNF-alpha level, caspase-3 and c-fos expressions and apoptotic cell numbers (all <0.05), but there was no significant difference between last two groups (all >0.05). Propofol 29-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 34402265-5 2021 Compared with surgery group, propofol group and surgery with propofol group showed increased TNF-alpha level, caspase-3 and c-fos expressions and apoptotic cell numbers (all <0.05), but there was no significant difference between last two groups (all >0.05). Propofol 61-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 34148507-10 2021 A significant increase in brain-derived neurotrophic factor and c-fos was found in DMF-treated rats compared with the CCH group (P<0.001). Dimethyl Fumarate 83-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 34148507-10 2021 A significant increase in brain-derived neurotrophic factor and c-fos was found in DMF-treated rats compared with the CCH group (P<0.001). 1-acetyl-2-(coumariniminecarboxamide-3-yl)hydrazine 118-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 34203104-7 2021 Our results showed that the high NAC dose stimulated cFOS expression in the NAcc, and that this effect was suppressed in the presence of MTEP, thus suggesting the implication of mGluR5. Acetylcysteine 33-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-57 34203104-7 2021 Our results showed that the high NAC dose stimulated cFOS expression in the NAcc, and that this effect was suppressed in the presence of MTEP, thus suggesting the implication of mGluR5. 3-((2-methyl-1,3-thiazol-4-yl)ethynyl)piperidine 137-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-57 34203104-8 2021 Additionally, high doses could attenuate the ethanol-induced increase in cFOS-expression in the NAcc, probably due to a phenomenon based on the long-term depression of the MSNs. Ethanol 45-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-77 35581381-9 2022 Interestingly, SL c-Fos expression was reduced at recall concomitant with impairment in cocaine-seeking behavior, suggesting that SL neurons may also facilitate cocaine-memory retrieval by inhibiting non-engram neuronal activity. Cocaine 161-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 35513168-8 2022 Mechanistically, oxLDL acts through mtROS to enhance transcription activity of c-Fos to facilitate the expression of LOX-1, exerting a feedforward mechanism with oxLDL to increase lipid uptake and propel VSMC-derived foam cell formation and atherogenesis. Mometasone Furoate 197-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 35378111-7 2022 Water-receiving animals had less c-Fos expression in the anterior basolateral amygdala than others groups. Water 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 34253253-0 2021 Assessing the treatment of cannabidiolic acid methyl ester: a stable synthetic analogue of cannabidiolic acid on c-Fos and NeuN expression in the hypothalamus of rats. cannabidiolic acid 91-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 35508252-6 2022 Eugenol promoted the secretion of GLP-1 into the blood, increased GLP-1 receptor (GLP-1R) expression in the duodenum, liver, arcuate nucleus (ARC) and paraventricular nucleus (PVN), increased c-fos expression in the nucleus tractus solitarii (NTS), and promoted ZO-1 and occludin expression in duodenum. Eugenol 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 192-197 35583808-8 2022 Sulfation of the TRPV1 receptor resulted in vagus nerve activation and promoted the c-fos and ChAT in the nucleus tractus solitaries with berberine. Berberine 138-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 35473765-8 2022 Females displayed greater recruitment (Fos expression) of the dorsal hippocampus than males, particularly in cold water. Water 114-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 35618945-11 2022 The muscimol response was compounded with the decreased levels of BDNF and c-Fos in the PFC and the hippocampus. Muscimol 4-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 35618945-12 2022 Thus, the GABA-A receptor mechanism may mediate the inhibitory effect of this probiotic mixture on stress-induced amnesia, which may be associated with the PFC and hippocampal BDNF/c-Fos signaling changes. gamma-Aminobutyric Acid 10-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 181-186 35615566-10 2022 Intra-LHb injection of the ERbeta-selective agonist diarylprepionitrile (DPN) reduced expression of c-Fos (a neuronal activity marker) and anxiety-like behavior in OVX rats, but not in normal rats, as evidenced by increased time spent in EPM open areas and the MCM mirror chamber. intra-lhb 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 35615566-10 2022 Intra-LHb injection of the ERbeta-selective agonist diarylprepionitrile (DPN) reduced expression of c-Fos (a neuronal activity marker) and anxiety-like behavior in OVX rats, but not in normal rats, as evidenced by increased time spent in EPM open areas and the MCM mirror chamber. diarylprepionitrile 52-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 35615566-10 2022 Intra-LHb injection of the ERbeta-selective agonist diarylprepionitrile (DPN) reduced expression of c-Fos (a neuronal activity marker) and anxiety-like behavior in OVX rats, but not in normal rats, as evidenced by increased time spent in EPM open areas and the MCM mirror chamber. dpn 73-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 35129799-13 2022 Meanwhile, the intracellular ROS level, the proportion of c-Fos+ cells, apoptosis rate, and nuclear translocation of NF-kappaB protein after treatment with BHBA were significantly decreased when compared with that in low glucose cells. 3-Hydroxybutyric Acid 156-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 35227807-0 2022 Oral exposure to high-dose ethanol for 28 days in rats reduces neural stem cells and immediate nascent neural progenitor cells as well as FOS-expressing newborn granule cells in adult hippocampal neurogenesis. Ethanol 27-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-141 35227807-8 2022 EtOH exposure (16%) also decreased the number of FOS+ granule cells, suggesting that synaptic plasticity was suppressed; concurrent upregulation of glutamate receptor/transporter genes may have occurred as a compensatory response against suppressed synaptic plasticity. Ethanol 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 35624974-7 2022 The analysis of c-Fos expression in auditory-related brain areas revealed that GJG significantly reduced the salicylate-induced increase in the number of c-Fos-expressing cells in the auditory cortices, inferior colliculus, and dorsal cochlear nucleus. Salicylates 109-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 35624974-7 2022 The analysis of c-Fos expression in auditory-related brain areas revealed that GJG significantly reduced the salicylate-induced increase in the number of c-Fos-expressing cells in the auditory cortices, inferior colliculus, and dorsal cochlear nucleus. Salicylates 109-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 35531473-7 2022 The expressions of p-CREB and c-fos proteins in the prefrontal cortex and nucleus accumbens of rats in the atimezazole group were higher than those in the model group. atimezazole 107-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 35531473-8 2022 In conclusion, atipamezole preconditioning can reduce cognitive dysfunction in aged rats after general anesthesia, and its mechanism may be related to inhibiting hippocampal inflammatory reaction and improving protein expression levels of p-CREB and c-fos in related brain regions of aged rats. atipamezole 15-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 250-255 35449195-6 2022 We demonstrate that a relatively low dose of DCZ (0.1 mg/kg) supports excitatory DREADD-mediated cFos induction, DREADD-mediated inhibition of a central amygdala-dependent behavior, and DREADD-mediated inhibition of neuronal activity in a slice electrophysiology preparation. Deoxycytidine 45-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-101 35108500-10 2022 This effect of SA seems to be related to the higher number of c-Fos/TH+ neurons in the A1 and A2. 2-chloro-10-(4'(N-beta-hydroxyethyl)piperazinyl-1')acetylphenothiazine 15-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 35404776-0 2022 The 5-HT7 receptor antagonist SB-269970 alleviates seizure activity and downregulates hippocampal c-Fos expression in pentylenetetrazole-induced kindled rats. SB 269970 30-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 35404776-0 2022 The 5-HT7 receptor antagonist SB-269970 alleviates seizure activity and downregulates hippocampal c-Fos expression in pentylenetetrazole-induced kindled rats. Pentylenetetrazole 118-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 35404776-12 2022 In addition, SB-269970 reduced the number of c-Fos positive cells in hippocampal CA1 area. SB 269970 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 35600629-12 2022 At this stage, brain function, specifically brain mitochondrial function, is not disturbed so it could be that the action of PBM in the mitochondria may not be detectable using the analysis of CCO activity and c-Fos protein expression. pbm 125-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 210-215 35409327-5 2022 Formalin was injected in the hindpaw both for behavioral nociceptive evaluation and the immunodetection of Fos expression in the spinal cord. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 35409327-6 2022 Hydrocephalic rats presented with a higher expression of TH at the LC, of TH and DBH at the spinal dorsal horn along with decreased nociceptive behavioral responses in the second (inflammatory) phase of the formalin test, and formalin-evoked Fos expression at the spinal dorsal horn. Formaldehyde 226-234 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 242-245 35571405-12 2022 Besides, TQDD inhibited p38MAPK pathway in colon and lung tissues, as well as reduced ATF2, c-jun, and c-fos expressions in colon and lung tissues. tqdd 9-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 35181484-10 2022 In the DAT-RNAi group, during the period of propofol anesthesia, the beta wave frequencies increased, the theta wave frequencies decreased, and the expression of c-fos protein in PFC increased and during the recovery from propofol anesthesia, the alpha wave and beta wave frequencies were increased (P < 0.05). Propofol 222-230 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 35169865-11 2022 In addition, ICA inhibited the protein expression of receptor activator of nuclear factor kappa-Beta ligand (RANKL), receptor activator of nuclear factor kappa-B (RANK), p38, ERK, c-Fos and nuclear factor of activated T cells 1 (NFATc1) in the femur of rats treated with TAA. icariin 13-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 180-185 35304863-7 2022 The duration of pain-related behaviors was prolonged in both phases I (0-5 min) and II (10-60 min) and the interphase; and the number of c-Fos-immunoreactive nuclei increased in laminae I-II, III-IV, and V-VI at the spinal segments L3-L5 on the side ipsilateral to the formalin injection, and these factors were significantly and positively correlated. Formaldehyde 269-277 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 35239670-0 2022 Clozapine prevented social interaction deficits and reduced c-Fos immunoreactivity expression in several brain areas of rats exposed to acute restraint stress. Clozapine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 35239670-6 2022 Conversely, pretreatment with clozapine, prevented social withdrawal and reduced c-Fos expression. Clozapine 30-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 35229934-0 2022 Fos-expressing neuronal ensembles in rat infralimbic cortex encode initial and maintained oxycodone seeking in rats. Oxycodone 90-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 35082908-3 2022 Moreover, ghrelin intervention increased PTEN expression level and reduced ERK, c-jun, and c-Fos expression level; in vivo experiments demonstrated that ghrelin intervention reduces mast memory expression and increases cardiomyocyte surface area, PTEN expression level, ERK, c-jun, c-Fos expression level, and cell surface area, while ERK blockade suppresses mast gene expression and reduces cell surface area. Ghrelin 10-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 35018823-12 2022 administration of PEG with significant induction of Fos-immunoreactive (-ir) neurons. Polyethylene Glycols 18-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 35126714-0 2022 Erratum: Metoprolol and bisoprolol ameliorate hypertrophy of neonatal rat cardiomyocytes induced by high glucose via the PKC/NF-kappaB/c-fos signaling pathway. Metoprolol 9-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 35126714-0 2022 Erratum: Metoprolol and bisoprolol ameliorate hypertrophy of neonatal rat cardiomyocytes induced by high glucose via the PKC/NF-kappaB/c-fos signaling pathway. Bisoprolol 24-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 35126714-0 2022 Erratum: Metoprolol and bisoprolol ameliorate hypertrophy of neonatal rat cardiomyocytes induced by high glucose via the PKC/NF-kappaB/c-fos signaling pathway. Glucose 105-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 35267900-9 2022 In addition, sucrose and naltrexone altered c-Fos IR in an interactive fashion in brain regions known to be involved in ingestion behavior. Sucrose 13-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 35267900-9 2022 In addition, sucrose and naltrexone altered c-Fos IR in an interactive fashion in brain regions known to be involved in ingestion behavior. Naltrexone 25-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 35051433-9 2022 In addition, the MS-group presented 180%, 137%, 170%, and 138% higher c-Fos-ir neurons for the ether exposure in the LC, PVN, MPA, and MeA, respectively. Ether 95-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 35185524-4 2022 Methods: We examined the effect of acute levodopa treatment on striatal c-Fos expression in LID using D1-Cre PD rats with dyskinetic symptoms induced by chronic levodopa administration. Levodopa 41-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 35185524-7 2022 Results: Striatal D1 + neurons in LID rats showed increased expression of c-Fos, a widely used marker for neuronal activation, following levodopa injection. Levodopa 137-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 35205303-0 2022 Mild Hypophagia and Associated Changes in Feeding-Related Gene Expression and c-Fos Immunoreactivity in Adult Male Rats with Sodium Valproate-Induced Autism. Valproic Acid 125-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 35082908-3 2022 Moreover, ghrelin intervention increased PTEN expression level and reduced ERK, c-jun, and c-Fos expression level; in vivo experiments demonstrated that ghrelin intervention reduces mast memory expression and increases cardiomyocyte surface area, PTEN expression level, ERK, c-jun, c-Fos expression level, and cell surface area, while ERK blockade suppresses mast gene expression and reduces cell surface area. Ghrelin 10-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 282-287 35082908-3 2022 Moreover, ghrelin intervention increased PTEN expression level and reduced ERK, c-jun, and c-Fos expression level; in vivo experiments demonstrated that ghrelin intervention reduces mast memory expression and increases cardiomyocyte surface area, PTEN expression level, ERK, c-jun, c-Fos expression level, and cell surface area, while ERK blockade suppresses mast gene expression and reduces cell surface area. Ghrelin 153-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 35082908-3 2022 Moreover, ghrelin intervention increased PTEN expression level and reduced ERK, c-jun, and c-Fos expression level; in vivo experiments demonstrated that ghrelin intervention reduces mast memory expression and increases cardiomyocyte surface area, PTEN expression level, ERK, c-jun, c-Fos expression level, and cell surface area, while ERK blockade suppresses mast gene expression and reduces cell surface area. Ghrelin 153-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 282-287 35095514-7 2021 Results: c-Fos and CBS co-expressed the most positive neurons after 1 h of restraint and immersion, followed by 3 h, and the least was at 0 h. After injection of different concentrations of NaHS into the PVN, gastric motility and gastric acid secretion in rats were significantly inhibited and promoted, respectively (p < 0.01); however, injection of normal saline, D-AP5, and PDTC did not cause any significant change (p > 0.05). 2-amino-4-oxo-5-phosphonopentanoic acid 366-371 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 35095514-7 2021 Results: c-Fos and CBS co-expressed the most positive neurons after 1 h of restraint and immersion, followed by 3 h, and the least was at 0 h. After injection of different concentrations of NaHS into the PVN, gastric motility and gastric acid secretion in rats were significantly inhibited and promoted, respectively (p < 0.01); however, injection of normal saline, D-AP5, and PDTC did not cause any significant change (p > 0.05). pyrrolidine dithiocarbamic acid 377-381 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 35095514-9 2021 Conclusion: There are neurons co-expressing CBS and c-Fos in the PVN, and the injection of NaHS into the PVN can inhibit gastric motility and promote gastric acid secretion in rats. sodium bisulfide 91-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 2514685-1 1989 Cyclosporin A, immunosuppressive agent, reversibly blocks the mitogenic effect of prolactin in rat lymphoma Nb-2 cells and removal from the medium leads to a rapid and transient induction of c-fos mRNA. Cyclosporine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 191-196 34986829-4 2022 T-5224 is a specific inhibitor of c-Fos/AP-1, that controls the gene expression of many proinflammatory cytokines. 3-(5-(4-(cyclopentyloxy)-2-hydroxybenzoyl)-2-((3-hydroxy-1,2-benzisoxazol-6-yl)methoxy)phenyl)propionic acid 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 34979902-9 2022 TNC neuroinflammation with increases in cytokines, CGRP and c-Fos and activation of the NF-kappaB pathway was observed, and these changes were alleviated by ibuprofen. Ibuprofen 157-166 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 34980778-9 2022 Mechanistically, curcumin + HH suppressed protein expression of stromal cell-derived factor-1 (SDF-1), CXC chemokine receptor 4 (CXCR4), p-Akt, and c-fos while enhancing protein expression of nerve growth factor (NGF) in the dorsal root ganglia (DRG) of model rats. Curcumin 17-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 35435121-6 2022 The C-Fos-positive reaction induced by SPS was mainly localized in neurons of the spinal dorsal horn, in which the C-Fos-positive cell density and its protein and mRNA levels were upregulated on SPS days 7 and 14; these changes were statistically significant in the SPS + EphrinB1-Fc group compared with the SPS alone group. Sodium phenolsulfonate 39-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 35435121-6 2022 The C-Fos-positive reaction induced by SPS was mainly localized in neurons of the spinal dorsal horn, in which the C-Fos-positive cell density and its protein and mRNA levels were upregulated on SPS days 7 and 14; these changes were statistically significant in the SPS + EphrinB1-Fc group compared with the SPS alone group. Sodium phenolsulfonate 39-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 35435121-6 2022 The C-Fos-positive reaction induced by SPS was mainly localized in neurons of the spinal dorsal horn, in which the C-Fos-positive cell density and its protein and mRNA levels were upregulated on SPS days 7 and 14; these changes were statistically significant in the SPS + EphrinB1-Fc group compared with the SPS alone group. Sodium phenolsulfonate 308-311 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 35435121-6 2022 The C-Fos-positive reaction induced by SPS was mainly localized in neurons of the spinal dorsal horn, in which the C-Fos-positive cell density and its protein and mRNA levels were upregulated on SPS days 7 and 14; these changes were statistically significant in the SPS + EphrinB1-Fc group compared with the SPS alone group. Sodium phenolsulfonate 308-311 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 2532558-4 1989 In unilaterally lesioned animals, L-dopa and the D1-selective agonists SKF 38393 and CY 208-243 produce contralateral rotation and induction of the nuclear proto-oncogene c-fos in the lesioned striatum. Levodopa 34-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-176 2532558-4 1989 In unilaterally lesioned animals, L-dopa and the D1-selective agonists SKF 38393 and CY 208-243 produce contralateral rotation and induction of the nuclear proto-oncogene c-fos in the lesioned striatum. Cysteine 85-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-176 2532558-5 1989 D-Amphetamine induces both ipsilateral rotation and c-fos activation in the intact striatum. Dextroamphetamine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 2532558-8 1989 Second, D1-dopamine agonists produce activation of striatal c-fos even if rotation is prevented by an anaesthetic. Dopamine 11-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 2514685-2 1989 Activators of protein kinase C, such as TPA, mellitin and phospholipase C and the calcium ionophore, A23187, induced c-fos mRNA in the presence or absence of cyclosporin A. Tetradecanoylphorbol Acetate 40-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 2571413-5 1989 Within 1 h of treatment, mature mRNA transcripts for c-fos were induced 6-fold, returning to control levels by 2 h. Other genes showed transiently elevated transcript levels after 2 h (c-Ha-ras, c-Ki-ras) or after 8 h (c-myc,c-myb, beta-actin, and Mr 70,000 heat shock protein) of testosterone replacement. Testosterone 281-293 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 2514685-2 1989 Activators of protein kinase C, such as TPA, mellitin and phospholipase C and the calcium ionophore, A23187, induced c-fos mRNA in the presence or absence of cyclosporin A. Calcium 82-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 2514685-2 1989 Activators of protein kinase C, such as TPA, mellitin and phospholipase C and the calcium ionophore, A23187, induced c-fos mRNA in the presence or absence of cyclosporin A. Calcimycin 101-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 2504783-11 1989 However, following 15 minutes of electrical stimulation of hindlimb cortex, Fos was expressed 4 hours later in patches of granule cell nuclei in Cop P and P. These patches of Fos immunostained granule cells occurred in similar locations in Cop P to the patches of highest glucose metabolism observed with the 2DG method. Glucose 272-279 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 2615190-7 1989 By contrast, cisplatin increased renal c-fos mRNA. Cisplatin 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 2558926-5 1989 Furthermore, administration of dexamethasone 2 h prior to 1-h immobilization attenuated the stress-enhanced immunostaining for the c-fos-like protein. Dexamethasone 31-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-136 2572704-5 1989 Carbachol induced accumulation of mRNA for c-fos and the other TIS genes. Carbachol 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 2503513-0 1989 Calcium induction of the mRNAs for prolactin and c-fos is independent of protein kinase C activity. Calcium 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 2503513-5 1989 Exposure of Ca2+-deprived GH3 cells to this ion in the presence of the Ca2+ channel modulator Bay K8644 yielded large increases in the mRNAs for both prolactin and c-fos. 3'-dGTP 26-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-169 2503513-7 1989 The latter observation was apparently not due simply to an inability of TPA to down-regulate the gene-regulatory activity of kinase C in intact GH3 cells, since this phorbol ester blocked the stimulation by platelet-derived growth factor of cellular levels of c-fos mRNA. Phorbol Esters 166-179 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 260-265 2507902-5 1989 Transforming ras protein activated an intracellular signal pathway, which led to the induction of 12-O-tetradecanoyl phorbol-13-acetate-responsive elements; activation was enhanced by coexpression of the proto-oncogene jun (encoding AP-1) and was further augmented by fos. Tetradecanoylphorbol Acetate 98-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 268-271 2505198-4 1989 Agents that interfered with the production of LTP (e.g. NMDA antagonists) also prevented c-fos induction. N-Methylaspartate 56-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 2504783-11 1989 However, following 15 minutes of electrical stimulation of hindlimb cortex, Fos was expressed 4 hours later in patches of granule cell nuclei in Cop P and P. These patches of Fos immunostained granule cells occurred in similar locations in Cop P to the patches of highest glucose metabolism observed with the 2DG method. Deoxyglucose 309-312 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 2739659-4 1989 FSH, serum, and phorbol esters individually stimulate c-fos in cultured Sertoli cells whereas platelet-derived growth factor, epidermal growth factor, and insulin-like growth factor-I have little affect. Phorbol Esters 16-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 2650909-5 1989 The results indicate that in the nodule DNA, c-myc and c-fos are hypomethylated in the sequence of CCGG while the c-Ha-ras shows hypomethylation in the alternating GCGC sequence. ccgg 99-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 2501508-2 1989 Induction of c-fos by epidermal growth factor, A23187, dBcAMP, or TPA in the same cells is not affected. Calcimycin 47-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 2501508-2 1989 Induction of c-fos by epidermal growth factor, A23187, dBcAMP, or TPA in the same cells is not affected. Bucladesine 55-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 2501508-2 1989 Induction of c-fos by epidermal growth factor, A23187, dBcAMP, or TPA in the same cells is not affected. Tetradecanoylphorbol Acetate 66-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 2494994-0 1989 Transient increase in the c-fos mRNA level after change of culture condition from serum absence to serum presence and after cycloheximide addition in rat 3Y1 fibroblasts. Cycloheximide 124-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 2494994-2 1989 Subsequent serum deprivation followed by serum re-addition or subsequent cycloheximide addition caused a transient re-increase in the c-fos mRNA level. Cycloheximide 73-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-139 2538365-7 1989 In addition to AVP, phorbol dibutyrate (PDBu), an activator of protein kinase C (PKC), also stimulates the accumulation of c-fos protein although to a lesser extent than AVP. Phorbol 12,13-Dibutyrate 20-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 2538365-7 1989 In addition to AVP, phorbol dibutyrate (PDBu), an activator of protein kinase C (PKC), also stimulates the accumulation of c-fos protein although to a lesser extent than AVP. Phorbol 12,13-Dibutyrate 40-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 2538365-8 1989 The above data suggest that c-fos protein levels in smooth muscle cells are regulated by AVP and the hormonal effect may be mediated through PI turnover and DAG, IP3 and Ca2+ signals. Diglycerides 157-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 2538365-8 1989 The above data suggest that c-fos protein levels in smooth muscle cells are regulated by AVP and the hormonal effect may be mediated through PI turnover and DAG, IP3 and Ca2+ signals. Inositol 1,4,5-Trisphosphate 162-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 2850967-3 1988 An element centered at position -60 of the c-fos promoter, which encompasses a consensus cAMP response element (CRE), is sufficient to confer cAMP responsiveness to a herpes thymidine kinase/CAT fusion gene. Cyclic AMP 89-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 2546062-9 1989 Both 8-CPT-cAMP and EGF were also equally effective in causing a rapid and transient induction of c-fos and c-myc protooncogene mRNA levels when added to growth-arrested H4IIE cells while A23187, N-(Bu)2-cAMP, and 4 beta-phorbol 12-myristate 13-acetate were significantly less effective. 8-(4-Chlorophenylthio)-cAMP 5-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 2538812-9 1989 A possible role for the NGF-stimulated diacylglycerol is suggested by the inhibition of NGF-dependent c-fos induction by staurosporin, a potent inhibitor of protein kinase C. These results suggest that, like insulin, some of the cellular effects of NGF may be mediated by the phospholipase C-catalyzed hydrolysis of glycosylphosphatidylinositol. Diglycerides 39-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 2538812-9 1989 A possible role for the NGF-stimulated diacylglycerol is suggested by the inhibition of NGF-dependent c-fos induction by staurosporin, a potent inhibitor of protein kinase C. These results suggest that, like insulin, some of the cellular effects of NGF may be mediated by the phospholipase C-catalyzed hydrolysis of glycosylphosphatidylinositol. Staurosporine 121-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 2472150-10 1989 Nuclear runoff transcription of the proto-oncogenes c-src, c-fms, c-sis, N-ras, c-myc, and c-fos was observed in proliferating and retinoic acid-treated cells. Tretinoin 131-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 3144275-0 1988 Morphine activation of c-fos expression in rat brain. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 3144275-1 1988 The post-receptor mechanism of opiate action has been studied by examining the activation by morphine of the proto-oncogene c-fos and its encoded nucleoprotein pp55c-fos (FOS) in rat caudate-putamen, which is rich in the mu-type opiate receptor. Morphine 93-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-129 3144275-1 1988 The post-receptor mechanism of opiate action has been studied by examining the activation by morphine of the proto-oncogene c-fos and its encoded nucleoprotein pp55c-fos (FOS) in rat caudate-putamen, which is rich in the mu-type opiate receptor. Morphine 93-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-174 3144275-2 1988 Following an acute morphine treatment, c-fos mRNA levels in rat caudate-putamen were increased to maximum (420% of control level) at 45 minutes and returned to control levels at 90 minutes. Morphine 19-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 3144275-4 1988 Fos protein, detected by immunocytochemistry, was also increased 3 hours after morphine injection, in the caudate-putamen, but not in the olfactory tubercle, which does not have the mu-type opiate receptor. Morphine 79-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 3144275-5 1988 Upon activation of opiate receptors by morphine, the c-fos gene is activated and Fos protein may act as a signal transducer uniquely involved in the mechanism of opiate addiction at the level of gene regulation. Morphine 39-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 3144275-5 1988 Upon activation of opiate receptors by morphine, the c-fos gene is activated and Fos protein may act as a signal transducer uniquely involved in the mechanism of opiate addiction at the level of gene regulation. Morphine 39-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-84 3144275-5 1988 Upon activation of opiate receptors by morphine, the c-fos gene is activated and Fos protein may act as a signal transducer uniquely involved in the mechanism of opiate addiction at the level of gene regulation. Opiate Alkaloids 19-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 3144275-5 1988 Upon activation of opiate receptors by morphine, the c-fos gene is activated and Fos protein may act as a signal transducer uniquely involved in the mechanism of opiate addiction at the level of gene regulation. Opiate Alkaloids 19-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-84 2844568-1 1988 Stimulation of beta-adrenoreceptors in rat parotid acinar cells in vitro by the beta-adrenergic agonist isoproterenol induces steady-state levels of c-fos mRNA and c-fos protein in these cells. Isoproterenol 104-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 149-154 2844568-1 1988 Stimulation of beta-adrenoreceptors in rat parotid acinar cells in vitro by the beta-adrenergic agonist isoproterenol induces steady-state levels of c-fos mRNA and c-fos protein in these cells. Isoproterenol 104-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-169 2844568-2 1988 A dramatic increase in the steady-state levels of c-fos mRNA was observed at 60 min, followed by a decrease at 2 h with a second peak at 4 h. c-fos induction in rat parotid acinar cells in vitro seems to be mediated by cAMP. Cyclic AMP 219-223 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 2844568-2 1988 A dramatic increase in the steady-state levels of c-fos mRNA was observed at 60 min, followed by a decrease at 2 h with a second peak at 4 h. c-fos induction in rat parotid acinar cells in vitro seems to be mediated by cAMP. Cyclic AMP 219-223 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-147 3152601-0 1988 Deregulation and overexpression of c-fos proto-oncogene in rat renal cell-lines and primary tumors induced by dimethylnitrosamine. Dimethylnitrosamine 110-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 3152601-1 1988 Rat renal mesenchymal tumors induced by the chemical carcinogen N-dimethylnitrosamine (DMN) and cell-lines derived from kidneys of rats after DMN treatment were found to express abnormal steady state levels of c-fos RNA. Dimethylnitrosamine 64-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 210-215 3152601-1 1988 Rat renal mesenchymal tumors induced by the chemical carcinogen N-dimethylnitrosamine (DMN) and cell-lines derived from kidneys of rats after DMN treatment were found to express abnormal steady state levels of c-fos RNA. Dimethylnitrosamine 87-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 210-215 3152601-1 1988 Rat renal mesenchymal tumors induced by the chemical carcinogen N-dimethylnitrosamine (DMN) and cell-lines derived from kidneys of rats after DMN treatment were found to express abnormal steady state levels of c-fos RNA. Dimethylnitrosamine 142-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 210-215 3141795-4 1988 We report here that estradiol causes a very rapid induction of the mRNA for the cellular oncogene c-fos in immature rat uterus. Estradiol 20-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 3141795-5 1988 Steady state levels of c-fos mRNA reach a maximum 3 h after 17 beta-estradiol administration and slowly return to low basal levels in 15 h. Dexamethasone, progesterone, and 5 alpha-dihydrotestosterone had no effect on uterine c-fos mRNA expression. Estradiol 60-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 3141795-5 1988 Steady state levels of c-fos mRNA reach a maximum 3 h after 17 beta-estradiol administration and slowly return to low basal levels in 15 h. Dexamethasone, progesterone, and 5 alpha-dihydrotestosterone had no effect on uterine c-fos mRNA expression. Dexamethasone 140-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 3141795-5 1988 Steady state levels of c-fos mRNA reach a maximum 3 h after 17 beta-estradiol administration and slowly return to low basal levels in 15 h. Dexamethasone, progesterone, and 5 alpha-dihydrotestosterone had no effect on uterine c-fos mRNA expression. Progesterone 155-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 3141795-6 1988 The induction of c-fos mRNA by estrogen was unaffected by the protein synthesis inhibitor puromycin but completely abolished by the RNA synthesis inhibitor actinomycin D. Puromycin 90-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 3141795-6 1988 The induction of c-fos mRNA by estrogen was unaffected by the protein synthesis inhibitor puromycin but completely abolished by the RNA synthesis inhibitor actinomycin D. Dactinomycin 156-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 3174663-1 1988 Administration of the convulsants pentylenetetrazole (Metrazole) or picrotoxin to rats caused a dramatic increase in mRNAs of four putative transcription factor genes, zif/268, c-jun, jun-B, and c-fos, in neurons of the hippocampus and dentate gyrus, as well as other areas of the cerebral cortex, including pyriform cortex and cingulate cortex. Pentylenetetrazole 54-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-200 2850250-5 1988 The kinetics of c-fos-driven CAT activity in response to TSH and cyclic AMP inducers were similar, and stimulation was reversible. Cyclic AMP 65-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 2850250-6 1988 These data therefore provide the first evidence that TSH and cyclic AMP stimulate the activity of the c-fos promoter in FRTL5 cells. Cyclic AMP 61-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 3134246-4 1988 The removal of TSA induced a rapid transient increase in the transcription of c-fos and the cells required 15 h to enter the S phase after release. trichostatin A 15-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 3136322-0 1988 Calcium and growth factor pathways of c-fos transcriptional activation require distinct upstream regulatory sequences. Calcium 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 3136322-1 1988 Transcription of the c-fos proto-oncogene is rapidly induced in the rat pheochromocytoma PC12 cell line by a wide variety of stimuli, including polypeptide growth factors, phorbol esters, and calcium ion fluxes. Phorbol Esters 172-186 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 3136322-1 1988 Transcription of the c-fos proto-oncogene is rapidly induced in the rat pheochromocytoma PC12 cell line by a wide variety of stimuli, including polypeptide growth factors, phorbol esters, and calcium ion fluxes. Calcium 192-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 3136322-4 1988 Calcium activation of c-fos transcription is dependent on a DNA element located approximately 60 base pairs upstream of the transcription start site. Calcium 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 3136322-5 1988 This region is highly conserved between human, mouse, and chicken c-fos genes and contains a sequence that resembles the consensus for a cyclic AMP response element. Cyclic AMP 137-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 3136322-7 1988 The dyad symmetry element is, however, an essential cis-acting sequence for c-fos inducibility by nerve growth factor, epidermal growth factor, fibroblast growth factor, and the phorbol ester 12-O-tetradecanoyl phorbol-13-acetate. phorbol ester 12-o-tetradecanoyl phorbol-13-acetate 178-229 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 2837462-4 1988 In rat thymic lymphocytes, the increase in the level of c-fos RNA induced by the combination of 12-O-tetradecanoylphorbol 13-acetate and ionomycin was unaffected by inhibition of the antiport with 5-(N-ethyl-N-propyl)amiloride. Tetradecanoylphorbol Acetate 96-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 2837462-4 1988 In rat thymic lymphocytes, the increase in the level of c-fos RNA induced by the combination of 12-O-tetradecanoylphorbol 13-acetate and ionomycin was unaffected by inhibition of the antiport with 5-(N-ethyl-N-propyl)amiloride. Ionomycin 137-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 3131879-4 1988 Moreover, 24-hour water deprivation resulted in Fos expression in paraventricular and supraoptic nuclei. Water 18-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 2769792-3 1989 In contrast, EGF, FGF, and TPA were equally effective in inducing accumulation of TIS8 and TIS28/c-fos mRNAs. Tetradecanoylphorbol Acetate 27-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 3147142-1 1988 Increases in intraneuronal free calcium result in the rapid, transient, induction of the fos and jun proto-oncogenes. Calcium 32-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 3147142-4 1988 In the brain, c-fos and c-jun may be induced by elevated neuronal activity such as occurs during pentylenetetrazole (PTZ) seizures. Pentylenetetrazole 97-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 3147142-4 1988 In the brain, c-fos and c-jun may be induced by elevated neuronal activity such as occurs during pentylenetetrazole (PTZ) seizures. Pentylenetetrazole 117-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 3147142-5 1988 The N-methyl-D-aspartate (NMDA) form of the glutamate receptor, which can directly gate calcium, plays a role in the induction of c-fos expression in PTZ seizures. N-Methylaspartate 4-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 3147142-5 1988 The N-methyl-D-aspartate (NMDA) form of the glutamate receptor, which can directly gate calcium, plays a role in the induction of c-fos expression in PTZ seizures. N-Methylaspartate 26-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 3147142-5 1988 The N-methyl-D-aspartate (NMDA) form of the glutamate receptor, which can directly gate calcium, plays a role in the induction of c-fos expression in PTZ seizures. Calcium 88-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 3147142-5 1988 The N-methyl-D-aspartate (NMDA) form of the glutamate receptor, which can directly gate calcium, plays a role in the induction of c-fos expression in PTZ seizures. Pentylenetetrazole 150-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 3147142-6 1988 In addition, NMDA can directly stimulate c-fos in the brain. N-Methylaspartate 13-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 3147142-9 1988 In this context Fos, Jun and the other immediate-early genes should be viewed as third messengers which are regulated by second messengers such as intracellular calcium. Calcium 161-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 2847942-3 1988 The induction of c-fos is specific since the expression of p53, a transformation-related gene, is not modulated by isoproterenol or 8-bromo-cAMP. Isoproterenol 115-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 2847942-3 1988 The induction of c-fos is specific since the expression of p53, a transformation-related gene, is not modulated by isoproterenol or 8-bromo-cAMP. 8-Bromo Cyclic Adenosine Monophosphate 132-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 3145095-3 1988 Here we report that a massive induction of c-fos protein is observed in dentate granule cells in four conditions that result in repetitive firing: localized seizure discharges; high frequency antidromic activation; orthodromic activation in the presence of iontophoresed bicuculline; and frequency potentiation. Bicuculline 271-282 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 2902626-8 1988 Such a factor, termed ATF (adenovirus transcription factor), has already been characterized and appears to have strong similarities to the transcriptional factor CREB (cAMP responsive element binding protein), which binds homologous sequences in cAMP responsive genes, such as somatostatin and c-fos. Cyclic AMP 168-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 294-299 3174663-1 1988 Administration of the convulsants pentylenetetrazole (Metrazole) or picrotoxin to rats caused a dramatic increase in mRNAs of four putative transcription factor genes, zif/268, c-jun, jun-B, and c-fos, in neurons of the hippocampus and dentate gyrus, as well as other areas of the cerebral cortex, including pyriform cortex and cingulate cortex. Pentylenetetrazole 34-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-200 3174663-1 1988 Administration of the convulsants pentylenetetrazole (Metrazole) or picrotoxin to rats caused a dramatic increase in mRNAs of four putative transcription factor genes, zif/268, c-jun, jun-B, and c-fos, in neurons of the hippocampus and dentate gyrus, as well as other areas of the cerebral cortex, including pyriform cortex and cingulate cortex. Picrotoxin 68-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-200 3282511-0 1988 Rapid and transient induction of c-fos, c-myc and c-Ha-ras in rat liver following glycine administration. Glycine 82-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 3282511-4 1988 The rapid rise and decline in the mRNA levels of c-fos, c-myc and c-Ha-ras in response to glycine is of significance because in response to a wide variety of growth stimuli, these proto-oncogenes exhibit a temporal sequence in their expression; for example, the expression of c-fos precedes that of c-myc, which in turn precedes the increased expression of c-Ha-ras. Glycine 90-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 3282511-4 1988 The rapid rise and decline in the mRNA levels of c-fos, c-myc and c-Ha-ras in response to glycine is of significance because in response to a wide variety of growth stimuli, these proto-oncogenes exhibit a temporal sequence in their expression; for example, the expression of c-fos precedes that of c-myc, which in turn precedes the increased expression of c-Ha-ras. Glycine 90-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 276-281 3382942-1 1988 The apparently non-specific accumulation of c-fos proto-oncogene mRNA was found in rat hippocampus as a result of injection of either glutamate, noradrenaline, or physiological saline. Glutamic Acid 134-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 3382942-1 1988 The apparently non-specific accumulation of c-fos proto-oncogene mRNA was found in rat hippocampus as a result of injection of either glutamate, noradrenaline, or physiological saline. Norepinephrine 145-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 3382942-1 1988 The apparently non-specific accumulation of c-fos proto-oncogene mRNA was found in rat hippocampus as a result of injection of either glutamate, noradrenaline, or physiological saline. Sodium Chloride 177-183 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 2850967-3 1988 An element centered at position -60 of the c-fos promoter, which encompasses a consensus cAMP response element (CRE), is sufficient to confer cAMP responsiveness to a herpes thymidine kinase/CAT fusion gene. Cyclic AMP 142-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 2850967-5 1988 Gel mobility shift assays with double-stranded oligonucleotides containing either the wild-type or mutated c-fos CRE sequence have demonstrated that binding occurs only to the wild-type CRE. double-stranded oligonucleotides 31-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 2850967-7 1988 Thus, regulation of the c-fos gene transcription appears to involve a mechanism common to many genes that respond to cAMP as a second message leading to cell growth and differentiation. Cyclic AMP 117-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 3130359-5 1988 c-fos expression was, however, significantly lower in TRb than TSb cells. Tri S Bond 63-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 3129127-7 1988 Generalized seizures evoked by injection of pentylenetetrazol produced a massive induction of fos protein(s) in the piriform and cingulate cortices as well as the dentate gyrus of rats. Pentylenetetrazole 44-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 2827679-0 1988 Angiotensin II induces expression of the c-fos gene through protein kinase C activation and calcium ion mobilization in cultured vascular smooth muscle cells. Calcium 92-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 2827679-2 1988 The doses of this agonist necessary for the increase in the c-fos mRNA level coincided with those for the phospholipase C-mediated hydrolysis of phosphoinositides. Phosphatidylinositols 145-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 2827679-3 1988 Moreover, protein kinase C-activating 12-O-tetradecanoylphorbol-13-acetate and Ca2+-ionophore A23187 increased the c-fos mRNA level in an additive manner. Tetradecanoylphorbol Acetate 38-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 2827679-3 1988 Moreover, protein kinase C-activating 12-O-tetradecanoylphorbol-13-acetate and Ca2+-ionophore A23187 increased the c-fos mRNA level in an additive manner. Calcimycin 94-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 3427445-5 1987 Like the increase in opioid peptide mRNA, the increase in c-fos mRNA began early in the period of seizure activity and could be blunted by maintaining the animals under anesthesia with the anticonvulsant sodium pentobarbital. Pentobarbital 204-224 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 3144696-1 1988 A rapid and transient induction of the c-fos transcript followed 4 hr later by long-term increase in the c-myc transcript was observed after disruption of the liver tissue with collagenase or EDTA perfusion and after in vitro detachment of the cell-sheet of liver cells in culture. Edetic Acid 192-196 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 3144696-2 1988 This increase of c-fos and c-myc transcripts could result from both an increased gene transcription and a stabilization of the corresponding mRNAs, as suggested by the effects of cycloheximide and actinomycin D. Cycloheximide 179-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 3144696-2 1988 This increase of c-fos and c-myc transcripts could result from both an increased gene transcription and a stabilization of the corresponding mRNAs, as suggested by the effects of cycloheximide and actinomycin D. Dactinomycin 197-210 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 2449626-1 1987 In primary cultures of rat cerebellar granule cells the activation of excitatory amino acid receptors by 1-glutamate enhances the steady state level of c-fos proto-oncogene messenger RNA. 1-glutamate 105-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-157 2449626-3 1987 Among the other excitatory amino acid agonists N-methyl-D-Aspartate (NMDA) and quisqualate also increased c-fos mRNA content, the latter however to a significantly lesser extent, while kainate failed to modify the basal level of c-fos expression. Excitatory Amino Acids 16-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 2449626-3 1987 Among the other excitatory amino acid agonists N-methyl-D-Aspartate (NMDA) and quisqualate also increased c-fos mRNA content, the latter however to a significantly lesser extent, while kainate failed to modify the basal level of c-fos expression. N-Methylaspartate 47-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 2449626-3 1987 Among the other excitatory amino acid agonists N-methyl-D-Aspartate (NMDA) and quisqualate also increased c-fos mRNA content, the latter however to a significantly lesser extent, while kainate failed to modify the basal level of c-fos expression. N-Methylaspartate 47-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 229-234 2449626-3 1987 Among the other excitatory amino acid agonists N-methyl-D-Aspartate (NMDA) and quisqualate also increased c-fos mRNA content, the latter however to a significantly lesser extent, while kainate failed to modify the basal level of c-fos expression. N-Methylaspartate 69-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 2449626-3 1987 Among the other excitatory amino acid agonists N-methyl-D-Aspartate (NMDA) and quisqualate also increased c-fos mRNA content, the latter however to a significantly lesser extent, while kainate failed to modify the basal level of c-fos expression. N-Methylaspartate 69-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 229-234 2449626-3 1987 Among the other excitatory amino acid agonists N-methyl-D-Aspartate (NMDA) and quisqualate also increased c-fos mRNA content, the latter however to a significantly lesser extent, while kainate failed to modify the basal level of c-fos expression. Quisqualic Acid 79-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 2449626-3 1987 Among the other excitatory amino acid agonists N-methyl-D-Aspartate (NMDA) and quisqualate also increased c-fos mRNA content, the latter however to a significantly lesser extent, while kainate failed to modify the basal level of c-fos expression. Quisqualic Acid 79-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 229-234 2856403-2 1987 In these basal conditions, the individual addition of TSH, insulin, insulin-like growth factor-I (IGF-I), phorbol 12-myristate 13-acetate (TPA), alpha 1-adrenergic agents, or A23187, increase c-myc and/or c-fos proto-oncogene expression. Tetradecanoylphorbol Acetate 106-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 205-210 3119359-2 1987 The expression of genes such as c-fos, c-myc, JE, KC, ornithine decarboxylase, and histone H3, analyzed by Northern blotting, increased in a cell-cycle dependent manner after colchicine treatment. Colchicine 175-185 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 2856403-3 1987 Under the same conditions, only the addition of TSH increased cAMP levels; 8-bromo-cAMP can increase c-myc or c-fos mRNA levels. 8-Bromo Cyclic Adenosine Monophosphate 75-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 3329719-0 1987 Forskolin and a tumor promoter are able to induce c-fos and c-myc expression in normal, but not in a v-ras-transformed rat thyroid cell line. Colforsin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 3329719-2 1987 c-fos and c-myc oncogenes expression was measured in these cells after addition of their specific growth factor TSH and after treatment with either forskolin, an activator of adenylate cyclase or with a tumor promoter, TPA. Colforsin 148-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 3329719-2 1987 c-fos and c-myc oncogenes expression was measured in these cells after addition of their specific growth factor TSH and after treatment with either forskolin, an activator of adenylate cyclase or with a tumor promoter, TPA. Tetradecanoylphorbol Acetate 219-222 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 3620471-1 1987 This study examined the effects of the mitogen, prolactin and the cell cycle inhibitors, cyclosporin A and neomycin sulfate, on expression of the proto-oncogenes c-fos and c-myc in the rat lymphoma Nb-2 cell line. Cyclosporine 89-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 3620471-1 1987 This study examined the effects of the mitogen, prolactin and the cell cycle inhibitors, cyclosporin A and neomycin sulfate, on expression of the proto-oncogenes c-fos and c-myc in the rat lymphoma Nb-2 cell line. Neomycin 107-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 3112583-2 1987 Expression of c-fos and c-myc are among the earliest genetic events induced in cultured fibroblast and phaeochromocytoma cell lines by various stimuli including growth factors, peptides and the intracellular second messengers diacylglycerol, cAMP and Ca2+. Diglycerides 226-240 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 3112583-2 1987 Expression of c-fos and c-myc are among the earliest genetic events induced in cultured fibroblast and phaeochromocytoma cell lines by various stimuli including growth factors, peptides and the intracellular second messengers diacylglycerol, cAMP and Ca2+. Cyclic AMP 242-246 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 3301843-7 1987 The phorbol ester phorbol 12-myristate 13-acetate (PMA) also induces c-myc and c-fos mRNAs without inducing DNA synthesis. Phorbol Esters 4-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 3301843-7 1987 The phorbol ester phorbol 12-myristate 13-acetate (PMA) also induces c-myc and c-fos mRNAs without inducing DNA synthesis. Tetradecanoylphorbol Acetate 18-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 3301843-7 1987 The phorbol ester phorbol 12-myristate 13-acetate (PMA) also induces c-myc and c-fos mRNAs without inducing DNA synthesis. Tetradecanoylphorbol Acetate 51-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 3301843-8 1987 However, the mechanism of this induction appears to be different from the insulin-induced induction since pretreatment of cells with PMA blocks only the PMA-mediated, not the insulin-mediated, induction of c-myc and c-fos. Tetradecanoylphorbol Acetate 133-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 216-221 3005310-0 1986 Thyrotropin and dibutyryl cyclic AMP increase levels of c-myc and c-fos mRNAs in cultured rat thyroid cells. Bucladesine 16-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 3620471-5 1987 However, the release of Nb-2 cells from a cyclosporin A or a neomycin sulfate block resulted in a rapid transient induction of c-fos which peaked at 0.5-1 h and declined rapidly thereafter. Cyclosporine 42-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 3620471-5 1987 However, the release of Nb-2 cells from a cyclosporin A or a neomycin sulfate block resulted in a rapid transient induction of c-fos which peaked at 0.5-1 h and declined rapidly thereafter. Neomycin 61-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 3536454-6 1987 Northern analysis with [32P]v-fos DNA revealed a major c-fos mRNA species (2.2 kilobases) induced by treatment of cells with IGF-I (100 ng/ml) or fetal calf serum (15%) for 45 min. Phosphorus-32 24-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 3325886-1 1987 c-fos mRNA accumulates to a level of 0.2% of cellular poly(A)-containing RNA 45 min after treatment of rat pheochromocytoma (PC12) cells with 1 mM barium chloride. Poly A 54-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 3325886-1 1987 c-fos mRNA accumulates to a level of 0.2% of cellular poly(A)-containing RNA 45 min after treatment of rat pheochromocytoma (PC12) cells with 1 mM barium chloride. barium chloride 147-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 3325886-6 1987 The c-fos protein synthesized in vitro appears to be phosphorylated by the cAMP-dependent protein kinase. Cyclic AMP 75-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 3330786-2 1987 Stimulation of c-fos expression by isoproterenol was inhibited by the beta-adrenergic antagonist propranolol. Isoproterenol 35-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 3330786-2 1987 Stimulation of c-fos expression by isoproterenol was inhibited by the beta-adrenergic antagonist propranolol. Propranolol 97-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 3330786-4 1987 In the heart of the rat, c-fos expression was also stimulated by the alpha-adrenergic agonist phenylephrine, histamine, and prostaglandin E1. Phenylephrine 94-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 3330786-4 1987 In the heart of the rat, c-fos expression was also stimulated by the alpha-adrenergic agonist phenylephrine, histamine, and prostaglandin E1. Histamine 109-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 3330786-4 1987 In the heart of the rat, c-fos expression was also stimulated by the alpha-adrenergic agonist phenylephrine, histamine, and prostaglandin E1. Alprostadil 124-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 3330786-5 1987 The histamine-induced expression of the c-fos gene was blocked by the histamine H1-receptor antagonist pyrilamine but not by H2-receptor antagonists ranitidine and cimetidine. Histamine 4-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 3330786-5 1987 The histamine-induced expression of the c-fos gene was blocked by the histamine H1-receptor antagonist pyrilamine but not by H2-receptor antagonists ranitidine and cimetidine. Pyrilamine 103-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 3502572-7 1987 Like c-fos, these TIS genes induced by NGF could also be superinduced by the combined administration of NGF and benzodiazepine. Benzodiazepines 112-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 5-10 3502572-8 1987 Elevated potassium ion, which leads to the induction of c-fos in PC-12 cells via activation of a voltage-dependent Ca2+ channel, also induces all TIS genes, with the notable exception of TIS 10. Potassium 9-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 2430291-0 1986 Barium modulates c-fos expression and post-translational modification. Barium 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 2430291-1 1986 The addition of exogenous barium ions to PC12 rat pheochromocytoma cells elicits a transient induction of the c-fos gene. Barium 26-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 2430291-3 1986 Thus, barium appears to enter the cell through a voltage-dependent calcium channel and, either directly or indirectly, interacts with calmodulin to stimulate c-fos expression. Barium 6-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 2430291-6 1986 The first comprises polypeptide growth factors and phorbol esters, which give rise to a c-fos protein that undergoes extensive post-translational modification. Phorbol Esters 51-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 2430291-7 1986 The second, which comprises depolarizing agents and barium ions, acts via calcium channels and yields a c-fos protein that undergoes less extensive post-translational modification. Barium 52-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 2430291-8 1986 These different forms of c-fos protein can be distinguished on the basis of their apparent molecular weights on sodium dodecyl sulfate/polyacrylamide gels. Sodium Dodecyl Sulfate 112-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 2430291-8 1986 These different forms of c-fos protein can be distinguished on the basis of their apparent molecular weights on sodium dodecyl sulfate/polyacrylamide gels. polyacrylamide 135-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 3014510-5 1986 In the presence of cycloheximide, the c-fos gene is superinduced and the increased level of c-fos mRNA persists for at least 24 hr. Cycloheximide 19-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 3014510-5 1986 In the presence of cycloheximide, the c-fos gene is superinduced and the increased level of c-fos mRNA persists for at least 24 hr. Cycloheximide 19-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 3005310-6 1986 At the single concentration tested (1 mM), Bt2cAMP also increased the level of c-fos mRNA. Bucladesine 43-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 3005310-7 1986 Hence, bTSH-stimulated mitogenesis in quiescent FRTL5 cells is associated with rapid, but short-lived, increases in the levels of the mRNAs of the proto-oncogenes, c-myc and c-fos. btsh 7-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 174-179 3005310-8 1986 Since bTSH is known to stimulate adenylate cyclase in these cells, and since the effect of TSH on c-myc and c-fos mRNAs is mimicked by Bt2cAMP, it is possible that these responses to bTSH are mediated, at least in part, by cAMP. Cyclic AMP 138-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 2997786-4 1985 cAMP, epidermal growth factor, the phorbol ester phorbol 12-myristate 13-acetate, and K+ depolarization also induce the fos gene. Cyclic AMP 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-123 4035354-0 1985 Superinduction of c-fos by nerve growth factor in the presence of peripherally active benzodiazepines. Benzodiazepines 86-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 2997786-4 1985 cAMP, epidermal growth factor, the phorbol ester phorbol 12-myristate 13-acetate, and K+ depolarization also induce the fos gene. Phorbol Esters 35-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-123 2997786-4 1985 cAMP, epidermal growth factor, the phorbol ester phorbol 12-myristate 13-acetate, and K+ depolarization also induce the fos gene. Tetradecanoylphorbol Acetate 49-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-123 34056932-7 2021 Caffeine injection also increased locomotor activity and c-Fos expression in the medial septum-vertical limb of the diagonal band of Broca (MS-VDB), one of the arousal regions of the basal forebrain. Caffeine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 33861997-0 2021 Cocaine-Induced Fos Expression in the Rat Brain: Modulation by Prior Delta9-Tetrahydrocannabinol Exposure During Adolescence and Sex-Specific Effects. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 33861997-0 2021 Cocaine-Induced Fos Expression in the Rat Brain: Modulation by Prior Delta9-Tetrahydrocannabinol Exposure During Adolescence and Sex-Specific Effects. Dronabinol 69-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 33861997-3 2021 Here we aimed at gaining a deeper understanding of the cellular activation patterns induced by cocaine as revealed by Fos imaging and how these patterns may change due to adolescent exposure to THC. Cocaine 95-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 33861997-8 2021 Cocaine-induced Fos activation was measured by immunohistochemistry as an index of cellular activation. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 33737065-2 2021 The offspring of dams, administered methylazoxymethanol acetate (MAM) intraperitoneally at gestational day 15, exhibit micrencephaly and anxiety-related behavior, such as hyperactivity in rearing and crossing behavior, alongside a distinct Fos expression profile in the basolateral (BLA) and central amygdala. Methylazoxymethanol Acetate 36-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 240-243 33737065-2 2021 The offspring of dams, administered methylazoxymethanol acetate (MAM) intraperitoneally at gestational day 15, exhibit micrencephaly and anxiety-related behavior, such as hyperactivity in rearing and crossing behavior, alongside a distinct Fos expression profile in the basolateral (BLA) and central amygdala. Methylazoxymethanol Acetate 65-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 240-243 33675933-9 2021 In addition, the RES treatment reduced the TH-immunoreactivity in the ventral tegmental area (VTA) and increased the c-Fos expression in the ventral-lateral periaqueductal gray (vlPAG), rostral ventral medulla (RVM) and dorsal raphe nucleus (DRN) after noxious stimuli induced by formalin. Reserpine 17-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 33887540-0 2021 The effect of amisulpride, olanzapine, quetiapine, and aripiprazole single administration on c-Fos expression in vasopressinergic and oxytocinergic neurons of the rat hypothalamic paraventricular nucleus. Aripiprazole 55-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 33887540-5 2021 The c-Fos was processed by avidin-biotin-peroxidase complex intensified with nickel-enhanced 3,3"-diaminobenzidine tetrahydrochloride. Nickel 77-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 33887540-5 2021 The c-Fos was processed by avidin-biotin-peroxidase complex intensified with nickel-enhanced 3,3"-diaminobenzidine tetrahydrochloride. 3,3'-DIAMINOBENZIDINE TETRAHYDROCHLORIDE 93-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 33753546-4 2021 These behavioral differences correlated with cocaine-induced activation of the RMTg, measured using both in vivo firing and cFos, while slice electrophysiological recordings from VTA-projecting RMTg neurons showed that, relative to low-avoiders, high-avoiders exhibited greater intrinsic excitability, greater transmission via calcium-permeable AMPA receptors (CP-AMPARs), and higher presynaptic glutamate release. Cocaine 45-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-128 34033853-7 2021 The high dose of propofol significantly increased c-fos expression in the ventrolateral preoptic nucleus (VLPO) sleep center and decreased the number of c-fos-immunoreactive neurons in wake-related systems including the tuberomammillary nucleus (TMN), perifornical nucleus (PeF), lateral hypothalamic nucleus (LH), ventrolateral periaqueductal gray (vPAG) and supramammillary region (SuM). Propofol 17-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 34033853-7 2021 The high dose of propofol significantly increased c-fos expression in the ventrolateral preoptic nucleus (VLPO) sleep center and decreased the number of c-fos-immunoreactive neurons in wake-related systems including the tuberomammillary nucleus (TMN), perifornical nucleus (PeF), lateral hypothalamic nucleus (LH), ventrolateral periaqueductal gray (vPAG) and supramammillary region (SuM). Propofol 17-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-158 33626390-8 2021 Furthermore, E177 reduced elevated levels of hippocampal c-fos protein expression in hippocampal tissues of PTZ-treated animals (all P < 0.05). Pentylenetetrazole 108-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 33953158-10 2021 By measuring the expression of immediate early genes (IEGs) as neuronal activity markers, we found that PBM treatment differentially regulated Arc and c-fos expression in the hippocampus and amygdala, two PTSD-related brain regions. pbm 104-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-156 3899692-1 1985 We have studied the expression of c-fos gene in rat hepatoma induced by DENA. Diethylnitrosamine 72-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 3899692-5 1985 In HTC cells the arrest of the cell cycle at early G1 phase by addition of sodium butyrate was accompanied by a strong increase of c-fos gene expression. Butyric Acid 75-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-136 3899692-6 1985 However the c-fos mRNA rapidly decreased after removal of sodium butyrate during the progression of the cells in the cell cycle and increased transiently when the cells entered again in G1 phase. Butyric Acid 58-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 34056932-9 2021 In addition, MAI treatment at HT7, compared with treatment at a location not corresponding to any traditional acupuncture point, reduced the caffeine-induced increase in c-Fos expression. Caffeine 141-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-175 32683756-0 2021 Fos-expressing neuronal ensemble in rat ventromedial prefrontal cortex encodes cocaine seeking but not food seeking in rats. Cocaine 79-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 33015936-7 2021 In rats trained on this procedure, this dose of alcohol also modulates expression of the IEGs c-Fos and Arc in brain regions known to modulate expression of alcohol interoceptive effects. Alcohols 48-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 33015936-7 2021 In rats trained on this procedure, this dose of alcohol also modulates expression of the IEGs c-Fos and Arc in brain regions known to modulate expression of alcohol interoceptive effects. Alcohols 157-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 33625560-7 2021 The DA + /5-HT-group had c-Fos activation in areas related to anti-predatory defensive behaviors, such as the ventromedial hypothalamic nucleus, premammillary nucleus, and periaqueductal gray. Dopamine 4-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 33625560-7 2021 The DA + /5-HT-group had c-Fos activation in areas related to anti-predatory defensive behaviors, such as the ventromedial hypothalamic nucleus, premammillary nucleus, and periaqueductal gray. Serotonin 10-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 33580994-7 2021 Mechanistically, ergothioneine reduced nuclear translocation of nuclear factor kappa B p65 and modulated p38 mitogen-activated protein kinase/c-Fos signaling. Ergothioneine 17-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-147 33454999-10 2021 Interestingly, the glucocorticoid Dexamethasone impairs early IL-6 signalling-induced mRNA expression of c-Fos and Egr1 and restrains neurite outgrowth. Dexamethasone 34-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 33559830-0 2021 MiR-101a-3p Attenuated Pilocarpine-Induced Epilepsy by Downregulating c-FOS. mir-101a-3p 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 33559830-0 2021 MiR-101a-3p Attenuated Pilocarpine-Induced Epilepsy by Downregulating c-FOS. Pilocarpine 23-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 33559830-12 2021 MiR-101a-3p was downregulated while c-FOS was increased in hippocampus tissues of Rat model of pilocarpine-induced epilepsy. Pilocarpine 95-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 33559830-16 2021 This study indicated that miR-101a-3p could attenuate pilocarpine-induced epilepsy by repressing c-Fos expression. mir-101a-3p 26-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 33559830-16 2021 This study indicated that miR-101a-3p could attenuate pilocarpine-induced epilepsy by repressing c-Fos expression. Pilocarpine 54-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 33571545-8 2021 We also observed activation (p<0.05) of c-Fos-sensitive neurons in the NTS and DVMN of exercised rats. dvmn 79-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 33789505-8 2021 Systemic OLZ decreased cisplatin-induced c-Fos immunofluorescence in the DVC and prevented cisplatin-induced reductions in circulating ghrelin levels. Olanzapine 9-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 33978481-10 2021 Conscious 24-27-week-old female OZRs had impaired baroreflex-mediated bradycardia, but fed blood glucose was modestly elevated (124.2+-5.2mg/dl) and phenylephrine-induced c-Fos expression in NTS was comparable to LZRs. Phenylephrine 149-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-176 33713748-0 2021 Insulin-Like Growth Factor-1 Inhibits Nitroglycerin-Induced Trigeminal Activation of Oxidative Stress, Calcitonin Gene-Related Peptide and c-Fos Expression. Nitroglycerin 38-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 33874962-11 2021 RESULTS: RTX injection induced mechanical allodynia and upregulated the protein expression of BDNF, TrkB.T1, ASIC3, TRAF6, nNOS, and c-Fos, as well as increased neuronal excitability in DRGs. resiniferatoxin 9-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 33600879-7 2021 Following FUS-triggered delivery, sham nanodroplets induced a 22.6 +- 21% increase in local c-Fos expression, whereas pentobarbital-loaded nanodroplets induced a 21.7 +- 13% decrease (n = 6). fusarubin 10-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 33897417-9 2021 Experimental validation analysis showed that CDG decreased the number of rotating laps and suppressed the levels of phosphorylated c-Jun, c-Fos, and JNK, as well as the number of TUNEL positive cells and the cleaved caspase-3 level in the nigrostriatal pathway. cdg 45-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 33592377-10 2021 Double immunofluorescence staining revealed that c-Fos-positive neurons in CSF-CN were significantly higher in the CIBP group than that in the sham group. cibp 115-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 33515270-7 2021 We also observed that 2C-C and 2C-P affected expression levels of the D1 dopamine receptor, D2 dopamine receptor, dopamine transporter, and phospho-dopamine transporter in the nucleus accumbens and the medial prefrontal cortex, and increased c-Fos immuno-positive cells in the nucleus accumbens. 2C-C 22-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 242-247 33515270-7 2021 We also observed that 2C-C and 2C-P affected expression levels of the D1 dopamine receptor, D2 dopamine receptor, dopamine transporter, and phospho-dopamine transporter in the nucleus accumbens and the medial prefrontal cortex, and increased c-Fos immuno-positive cells in the nucleus accumbens. 2C-P 31-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 242-247 33900100-6 2021 The results indicated that D-Trp8-gamma-MSH decreased the ingestion of the HFD and this effect is associated with the early development of the satiation process Moreover, the D-Trp8-gamma-MSH increased the accumulation of the alpha-MSH in the ARC and the c-Fos activity in the PVN. d-trp8-gamma-msh 27-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 255-260 33900100-6 2021 The results indicated that D-Trp8-gamma-MSH decreased the ingestion of the HFD and this effect is associated with the early development of the satiation process Moreover, the D-Trp8-gamma-MSH increased the accumulation of the alpha-MSH in the ARC and the c-Fos activity in the PVN. d-trp8-gamma-msh 175-191 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 255-260 33835816-7 2021 NAAB regulated the expression of Mgat4a, Gstm6, and Lpl, whereas AAPP modified the expression of Mgat4a, Jun, Fos, and Egr1. Ala-Ala-Pro-Pro 65-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 33596416-7 2021 Parallelly, sinomenine treatment significantly reduced the expression of various factors related to stress and inflammation, including p38 MAPK, NF-kappaB, c-fos, p-CAMKII, COX-2, p-CREB, TLR4 and IL-17A in DRG cells in vitro. sinomenine 12-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 33622703-7 2021 This was accompanied by altered GABA-A receptor subunit composition, increased dendritic complexity of apical dendrites of CA1 pyramidal neurons, and enhanced neuronal activation revealed by increased c-Fos-positive cell numbers within hippocampi of PNFlx treated animals in adulthood. pnflx 250-255 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 201-206 33789505-8 2021 Systemic OLZ decreased cisplatin-induced c-Fos immunofluorescence in the DVC and prevented cisplatin-induced reductions in circulating ghrelin levels. Cisplatin 23-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 33879516-14 2021 ICV tofogliflozin increased phosphorylation of AMPK and c-fos expression in the lateral hypothalamus. 6-((4-ethylphenyl)methyl)-3',4',5',6'-tetrahydro-6'-(hydroxymethyl)spiro(isobenzofuran-1(3H),2'-(2H)pyran)-3',4',5'-triol 4-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 33640680-3 2021 METHODS: This study compared neuronal activation, as reported by observable Fos expression in the IPN of nicotine-dependent female and male rats experiencing withdrawal. Nicotine 105-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 33657396-0 2021 Alteration of CYP2E1, DBN1, DNMT1, miRNA-335, miRNA-21, c-Fos and Cox-2 gene expression in prefrontal cortex of rats" offspring submitted to prenatal ethanol exposure during their neurodevelopment and the preventive role of nancocurcumin administration: A histological, ultrastructural and molecular study. Ethanol 150-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 32927465-35 2021 : In females (a-c; g-i), THC dose-dependently induced Fos immunoreactivity in several structures, as indicated by *symbol under bars that significantly differ from vehicle in that structure. Dronabinol 25-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 33581143-7 2021 Results showed that increased c-Fos expression in PPG neurons following LiCl and RES-but not Coc-is dependent on hindbrain 5-HT2C and 5-HT3 receptor signaling. Lithium Chloride 72-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 33581143-8 2021 Additionally, stressors that depend on 5-HT signaling to activate PPG neurons (i.e., LiCl and RES) increased c-Fos expression in 5-HT-expressing neurons within the caudal raphe (CR), specifically in the raphe magnus (RMg). Serotonin 39-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 33581143-8 2021 Additionally, stressors that depend on 5-HT signaling to activate PPG neurons (i.e., LiCl and RES) increased c-Fos expression in 5-HT-expressing neurons within the caudal raphe (CR), specifically in the raphe magnus (RMg). Lithium Chloride 85-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 33581143-8 2021 Additionally, stressors that depend on 5-HT signaling to activate PPG neurons (i.e., LiCl and RES) increased c-Fos expression in 5-HT-expressing neurons within the caudal raphe (CR), specifically in the raphe magnus (RMg). 4-phosphoerythronohydroxamic acid 94-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 33581143-8 2021 Additionally, stressors that depend on 5-HT signaling to activate PPG neurons (i.e., LiCl and RES) increased c-Fos expression in 5-HT-expressing neurons within the caudal raphe (CR), specifically in the raphe magnus (RMg). Serotonin 129-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 33786741-6 2021 Melatonin prevented the effect of experimental glaucoma on melanopsin-expressing RGC number, blue- and white light-induced pupil constriction, retina-olivary pretectal nucleus, and retina- suprachiasmatic nuclei communication, light-induced c-Fos expression in the suprachiasmatic nuclei, and alterations in the locomotor activity daily rhythm. Melatonin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 241-246 33136614-4 2021 Therefore, in this study, we evaluated the effects of doxapram in Fos (c-Fos) protein expression in the PAG and characterized its cardiorespiratory and behavioral effects on the elevated T maze and in the conditioned place aversion (CPA) paradigms. Doxapram 54-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 32927465-44 2021 Effects of THC doses on global network connectivity (THC-induced change in overall regional Fos cross-correlation) are shown in females (c) and males (d). Dronabinol 11-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 33136614-4 2021 Therefore, in this study, we evaluated the effects of doxapram in Fos (c-Fos) protein expression in the PAG and characterized its cardiorespiratory and behavioral effects on the elevated T maze and in the conditioned place aversion (CPA) paradigms. Doxapram 54-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 33136614-5 2021 Doxapram increased Fos expression in different columns of the PAG, increased respiratory frequency, decreased heart rate, and increased arterial pressure when injected via intravenous route. Doxapram 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 32927465-44 2021 Effects of THC doses on global network connectivity (THC-induced change in overall regional Fos cross-correlation) are shown in females (c) and males (d). Dronabinol 53-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 33754647-0 2021 Effects of Tacrolimus on c-Fos in Hippocampus and Memory Performances in Streptozotocin Model of Alzheimer"s Disease of Rats. Tacrolimus 11-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 33754647-0 2021 Effects of Tacrolimus on c-Fos in Hippocampus and Memory Performances in Streptozotocin Model of Alzheimer"s Disease of Rats. Streptozocin 73-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 33465355-6 2021 These neurochemical effects, which were replicated by the pERK1/2 inhibitor PD98059, likely underlie the reductions in c-Fos and caspase-3 levels as well as the anti-apoptotic effect of TLM in the IR model. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 76-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 33682265-9 2021 In the hippocampus, NIMP increased the expression of the neurotoxicity marker Fos and decreased the expression of neuroprotective Bdnf and antiapoptotic Bcl2l1. nimp 20-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 32418296-7 2021 Finally, posttraining naltrexone and exposure to the CS+ both induced significant expression of c-Fos in the CeA. Naltrexone 22-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 32418296-7 2021 Finally, posttraining naltrexone and exposure to the CS+ both induced significant expression of c-Fos in the CeA. Cesium 53-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 33422541-6 2021 The administration of levodopa increased c-Fos expression in a subpopulation of sensorimotor striatum neurons of dyskinetic rats, but not in non-dyskinetic rats. Levodopa 22-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 33833815-9 2021 By contrast, candesartan or candesartan + music therapy ameliorated the changes in retina cell apoptosis, reduction of neurotransmitters, increase in AII, and the expression of c-fos and BDNF. candesartan 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 177-182 33833815-9 2021 By contrast, candesartan or candesartan + music therapy ameliorated the changes in retina cell apoptosis, reduction of neurotransmitters, increase in AII, and the expression of c-fos and BDNF. candesartan 28-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 177-182 33422541-7 2021 The majority of the c-Fos+ neurons activated by levodopa in the striatum are positive for D5 receptor staining. Levodopa 48-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 33285237-0 2021 Morphine-Conditioned Placebo Analgesia in Female and Male Rats with Chronic Neuropathic Pain: c-Fos Expression in The Rostral Ventromedial Medulla. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 33285237-6 2021 We detected injury-specific c-Fos expression in the gracile nucleus and morphine-specific c-Fos expression in the serotonergic midline raphe nuclei and the caudal nuclei of the solitary tract. Morphine 72-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 33624592-9 2021 The results of ELISA and Western blotting showed that the expressions of c-Fos, NR4A1, p38MAPK and pp38MAPK increased significantly in cells with hypoxic exposure (P < 0.05); treatment with the serum containing Fuxinfang significantly reduced the expression levels of c-Fos, NR4A1 and p-p38MAPK in hypoxic HAECs in a concentration-dependent manner (P < 0.05). fuxinfang 211-220 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 33624592-11 2021 Fuxinfang improves hypoxic injuries of HAECs possibly by down-regulating the expression of c-Fos to inhibit NR4A1 expression and suppressing hypoxia-induced p38 phosphorylation. fuxinfang 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 33618583-0 2021 c-Fos is upregulated in the genital tubercle of DEHP-induced hypospadiac rats and the prepuce of patients with hypospadias. Diethylhexyl Phthalate 48-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 33618583-1 2021 This study aimed to investigate the expression of Fos proto-oncogene, AP-1 transcription factor subunit (c-Fos) in the genital tubercle (GT) of rats with di(2-ethylhexyl) phthalate (DEHP)-induced hypospadias and in the prepuce of patients with hypospadias compared with unaffected controls. Diethylhexyl Phthalate 154-180 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 33618583-1 2021 This study aimed to investigate the expression of Fos proto-oncogene, AP-1 transcription factor subunit (c-Fos) in the genital tubercle (GT) of rats with di(2-ethylhexyl) phthalate (DEHP)-induced hypospadias and in the prepuce of patients with hypospadias compared with unaffected controls. Diethylhexyl Phthalate 154-180 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 33618583-1 2021 This study aimed to investigate the expression of Fos proto-oncogene, AP-1 transcription factor subunit (c-Fos) in the genital tubercle (GT) of rats with di(2-ethylhexyl) phthalate (DEHP)-induced hypospadias and in the prepuce of patients with hypospadias compared with unaffected controls. Diethylhexyl Phthalate 182-186 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 33618583-1 2021 This study aimed to investigate the expression of Fos proto-oncogene, AP-1 transcription factor subunit (c-Fos) in the genital tubercle (GT) of rats with di(2-ethylhexyl) phthalate (DEHP)-induced hypospadias and in the prepuce of patients with hypospadias compared with unaffected controls. Diethylhexyl Phthalate 182-186 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 33618583-3 2021 Western blotting showed that c-Fos expression was increased in DEHP-induced hypospadiac male offspring. Diethylhexyl Phthalate 63-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 33618583-7 2021 c-Fos protein levels were higher in the GT of DEHP-induced rats than in that of control rats. Diethylhexyl Phthalate 46-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 33618583-9 2021 The expression of c-Fos was increased in both the GT of DEHP-induced hypospadiac rats and the prepuce of hypospadias patients. Diethylhexyl Phthalate 56-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 33347671-14 2021 Apelin-induced c-Fos expression in AP was partially attenuated by pretreatment of cholecystokinin-1 receptor antagonist lorglumide; whereas, it was completely abolished in vagotomized rats. lorglumide 120-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 32558119-11 2021 Ceftriaxone reduced Fos expression in regions sending projections to the NA core (prefrontal cortex, basolateral amygdala, ventral tegmental area) and specifically reduced Fos in prelimbic cortex and not infralimbic cortex neurons projecting to the NA core. Ceftriaxone 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 32558119-11 2021 Ceftriaxone reduced Fos expression in regions sending projections to the NA core (prefrontal cortex, basolateral amygdala, ventral tegmental area) and specifically reduced Fos in prelimbic cortex and not infralimbic cortex neurons projecting to the NA core. Ceftriaxone 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-175 32570285-8 2021 Incubation of oxycodone craving is accompanied with a time-dependent increase of Fos protein expression in both ventral and lateral OFC. Oxycodone 14-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-84 33169700-9 2021 The re-exposure to the alcohol-associated Context A reinstated alcohol seeking and increased Fos-positive cells in the dorsal hippocampus neurons (CA1, CA3, and Dentate Gyrus). Alcohols 23-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 33451281-3 2021 Acute treadmill running at low speed, regardless of exercise duration, significantly increased c-Fos expression in 5-HT neurons in the DRN compared with controls, whereas high-speed running significantly activated 5-HT neurons only at 60-min duration. Serotonin 115-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 32826755-7 2021 Moreover, morphine-induced effects on c-Fos (a marker of neuronal activity) in regions downstream of vlPAG, namely the rostral ventromedial medulla and lumbar spinal dorsal horn, were blunted in the WKY rats. Morphine 10-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 33574755-0 2020 Corydalis Saxicola Bunting Total Alkaloids Attenuate Walker 256-Induced Bone Pain and Osteoclastogenesis by Suppressing RANKL-Induced NF-kappaB and c-Fos/NFATc1 Pathways in Rats. Alkaloids 33-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 33574755-0 2020 Corydalis Saxicola Bunting Total Alkaloids Attenuate Walker 256-Induced Bone Pain and Osteoclastogenesis by Suppressing RANKL-Induced NF-kappaB and c-Fos/NFATc1 Pathways in Rats. walker 256 53-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 33708107-8 2020 Moreover, autophagy inhibitors, ROS scavenger, Ca2+ chelator and NF-kappaB inhibitor remarkably suppressed c-Fos and NFATc1 expressions. ros 32-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 33091429-10 2021 With correct use of landmarks for image warping, ABA produces similar results to manually drawn ROIs, results in no interobserver variability, and maintains c-Fos + pixel dimensions. alisol B 23-acetate 49-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-162 33451281-5 2021 c-Fos expression in 5-HT neurons in the DRN induced by the acute treadmill running for 30 or 60 min, but not 15 min, was positively correlated with the time spent on the open arms in the EPM and was negatively correlated with the immobility time in the FST. Serotonin 20-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 33214316-5 2021 Furthermore, rEPN lesions reduced cocaine-induced cFos activation by two-fold in the LHb and by a smaller proportion in the RMTg, while inactivation of the rEPN or the rEPN-LHb pathway attenuated cocaine avoidance behaviors measured by an operant runway task and by conditioned place aversion. Cocaine 34-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-54 33378960-9 2021 Subsequently, dioscin reduced the expressions of Ras, Cyclin D1, CDK4, c-Fos, PCNA and p-ERK to inhibit proliferation and migration of PASMCs, inhibited p-PI3K and p-AKT levels and increased Bax/Bcl2 ratio to promote cell apoptosis. dioscin 14-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 33420284-7 2021 Caffeine administered both IV and ICV increased neuronal activity, as measured by c-Fos-immunoreactivity within subregions of the hypothalamic area, previously implicated in regulating BAT thermogenesis. Caffeine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 33188843-6 2021 The ingestive behavior was not significantly affected by PHE treatment in the DR. CLO treatment increased Fos expression in the arcuate nucleus (ARC) and paraventricular nucleus of the hypothalamus (PVN) in rats that were allowed to eat during the experimental recording (AF group). Clonidine 82-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 33368748-7 2021 PQ + MB stimulated cAMP to reduce the production of protein kinase A (PKA) and inhibited the activation of other elements, such as brain-derived neurotrophic factor (BDNF), cAMP response element binding protein (CREB), phospho-CREB (p-CREB), immediate-early genes (IEGs)Arc, and c-Fos. Cyclic AMP 19-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 279-284 33188843-9 2021 However, double-labeled POMC/Fos cells were absent in the ARC of the WAF group, although an increase in Fos expression was observed in non-POMC cells after CLO injections in the WAF group. Clonidine 156-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 32978649-3 2021 The purpose of these studies was to examine if exposure of male Long-Evans rats to the synthetically derived predator odor 2,5-dihydro-2,4,5-trimethylthiazoline (TMT; a component of fox feces) would increase sweetened alcohol self-administration, potentially by facilitating transfer of salience towards cues, and alter neuronal response to alcohol as measured by the immediate early gene c-Fos. 2,5-dihydro-2,4,5-trimethylthiazoline 123-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 389-394 32978649-3 2021 The purpose of these studies was to examine if exposure of male Long-Evans rats to the synthetically derived predator odor 2,5-dihydro-2,4,5-trimethylthiazoline (TMT; a component of fox feces) would increase sweetened alcohol self-administration, potentially by facilitating transfer of salience towards cues, and alter neuronal response to alcohol as measured by the immediate early gene c-Fos. 2,5-dihydro-2,4,5-trimethylthiazoline 162-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 389-394 32978649-5 2021 In experiment 2, rats exposed to repeated TMT showed blunted basolateral amygdala c-Fos response to alcohol. Alcohols 100-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 33124673-9 2020 Stimulation of the AcbSh resulted in a decrease of c-fos mRNA expression in the LH and rostral VP, and stimulation of axonal terminals from the AcbSh to the rostral VP decreased c-fos mRNA expression only in the rostral VP. acbsh 144-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 178-183 33249495-5 2020 We also found the upregulation of serotonin and brain derived neurotrophic factor expression in the c-fos-positive areas of rats treated with blue light compared with those maintained in darkness. Serotonin 34-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 33276724-6 2020 The allodynia was assessed by mechanical and thermal thresholds, and central sensitization was assessed by expression of the phosphorylation of cyclic adenosine monophosphate (cAMP) response element-binding protein (CREB) at Serine 133(pCREB-S133) and c-Fos. Cyclic AMP 176-180 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 252-257 33276724-10 2020 Downregulated the mGluR5 with MPEP alleviated the allodynia and reduced the expression of CGRP, pCREB-S133, c-Fos, PSD, Syp and Syt-1 and synaptic transmission. 6-methyl-2-(phenylethynyl)pyridine 30-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 32931900-4 2020 To visualize the changes of AP neuron excitability after inhalation of MMA for 90 min, c-Fos protein expression was identified and quantified by immunohistochemical analysis. Methylmethacrylate 71-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 32931900-7 2020 In vagotomized rats inhaling MMA, markedly smaller number of Fos-ir cells was identified in the AP compared to that of rats inhaling MMA without vagotomy. Methylmethacrylate 29-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 32931900-8 2020 CONCLUSIONS: These results indicate that inhalation of MMA increases neuronal excitability in the AP, suggesting that vagal afferent inputs are involved in the induction mechanism of Fos-ir cells by MMA. Methylmethacrylate 55-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 183-186 32931900-8 2020 CONCLUSIONS: These results indicate that inhalation of MMA increases neuronal excitability in the AP, suggesting that vagal afferent inputs are involved in the induction mechanism of Fos-ir cells by MMA. Methylmethacrylate 199-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 183-186 33065316-7 2020 In the animals injected with emetine (0.5 and 1.0 mM), many c-Fos-like immunoreactive (Fos-ir) cells were observed in the area postrema and the NTS, while few Fos-ir cells were identified in the animals injected with saline. Emetine 29-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 33065316-7 2020 In the animals injected with emetine (0.5 and 1.0 mM), many c-Fos-like immunoreactive (Fos-ir) cells were observed in the area postrema and the NTS, while few Fos-ir cells were identified in the animals injected with saline. Emetine 29-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 33065316-7 2020 In the animals injected with emetine (0.5 and 1.0 mM), many c-Fos-like immunoreactive (Fos-ir) cells were observed in the area postrema and the NTS, while few Fos-ir cells were identified in the animals injected with saline. Emetine 29-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 33065316-8 2020 The average number of Fos-ir cells in the area postrema and the NTS was significantly larger in animals injected with emetine than in animals injected with saline. Emetine 118-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-25 33258348-0 2020 Protective effect of Chushizi (Fructus Broussonetiae) on acetaminophen-induced rat hepatitis by inhibiting the Toll-like receptor 3/c-Jun N-terminal kinase/c-jun/c-fos/janus protein tyrosine kinase/activators of transcription 3 pathway. Acetaminophen 57-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 33258348-9 2020 CONCLUSION: Our findings suggest that CSZ is a valid medicine to alleviate APAP-induced DILI, while its partial mechanism may regulate the TLR3/JNK/ c-jun/c-fos/JAK/STAT3 pathway. csz 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-160 32621298-8 2020 Training in the water maze increased c-Fos positive nuclei in the dentate gyrus of the hippocampus and in medial prefrontal cortex. Water 16-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 32628919-3 2020 The aim of the study was to examine if whether Nnat immunoreactivity is detectable in the NTS, and whether peripheral CCK-8S or BN cause c-Fos activation of Nnat neurons. cholecystokinin 8 118-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 33124673-9 2020 Stimulation of the AcbSh resulted in a decrease of c-fos mRNA expression in the LH and rostral VP, and stimulation of axonal terminals from the AcbSh to the rostral VP decreased c-fos mRNA expression only in the rostral VP. acbsh 19-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 33239732-5 2020 The predominant finding in rats receiving bilateral BST stimulation was a striking increase in c-Fos expression throughout much of the left hemisphere, which was not confined to the predefined regions of interest. growth hormone, bovine 52-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 32730891-16 2020 The degeneration of Purkinje neurons might have resulted from the overexpression of c-fos and increased oxidative stress in the cerebellum after the multiple dosing of TAEP. taep 168-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 33198169-5 2020 After having confirmed that the oral bouts induced by quinpirole 0.5 mg/kg were blocked by another 5-HT2C antagonist (6-chloro-5-methyl-1-[6-(2-methylpiridin-3-yloxy)pyridine-3-yl carbamoyl] indoline (SB 242084), 1 mg/kg), we mapped the changes in neuronal activity in numerous sub-territories of the basal ganglia using c-Fos expression. Quinpirole 54-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 321-326 33198169-6 2020 We found a marked increase of c-Fos expression in the subthalamic nucleus (STN) in combining quinpirole (0.5 mg/kg) with either SB 243213 or SB 242084. Quinpirole 93-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 33198169-6 2020 We found a marked increase of c-Fos expression in the subthalamic nucleus (STN) in combining quinpirole (0.5 mg/kg) with either SB 243213 or SB 242084. SB 243213 128-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 33198169-6 2020 We found a marked increase of c-Fos expression in the subthalamic nucleus (STN) in combining quinpirole (0.5 mg/kg) with either SB 243213 or SB 242084. 6-chloro-5-methyl-1-((2-(2-methylpyrid-3-yloxy)pyrid-5-yl)carbamoyl)indoline 141-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 32574519-0 2020 Changes in c-fos and p-CREB signaling following exposure to forced swim stress or exogenous corticosterone during morphine-induced place preference are dependent on glucocorticoid receptor in the basolateral amygdala. Corticosterone 92-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 32574519-0 2020 Changes in c-fos and p-CREB signaling following exposure to forced swim stress or exogenous corticosterone during morphine-induced place preference are dependent on glucocorticoid receptor in the basolateral amygdala. Morphine 114-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 32574519-4 2020 Our results showed that morphine-induced CPP could increase c-fos level and p-CREB/CREB ratio in all regions (except in the HIP). Morphine 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 32574519-6 2020 In the PFC, in addition to FSS, corticosterone could raise c-fos expression, which was blocked by RU38486. Corticosterone 32-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 32574519-6 2020 In the PFC, in addition to FSS, corticosterone could raise c-fos expression, which was blocked by RU38486. Mifepristone 98-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 32574519-7 2020 In conclusion, it seems that the intra-BLA administration of RU38486 differently modulates the effect of morphine-induced CPP on the expression of c-fos and p-CREB/CREB ratio in animals that underwent FSS or corticosterone administration. Mifepristone 61-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-152 32574519-7 2020 In conclusion, it seems that the intra-BLA administration of RU38486 differently modulates the effect of morphine-induced CPP on the expression of c-fos and p-CREB/CREB ratio in animals that underwent FSS or corticosterone administration. Morphine 105-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-152 32894768-6 2020 The 4V 2DG administration significantly increased the number of Pdyn (Dyn gene)-positive cells co-expressing fos in the paraventricular nucleus (PVN), but not in the ARC and supraoptic nucleus (SON), and the iv 2DG treatment significantly increased the number of fos- and Pdyn-co-expressing cells in the PVN and SON, but decreased it in the ARC. Deoxyglucose 7-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-112 32894768-6 2020 The 4V 2DG administration significantly increased the number of Pdyn (Dyn gene)-positive cells co-expressing fos in the paraventricular nucleus (PVN), but not in the ARC and supraoptic nucleus (SON), and the iv 2DG treatment significantly increased the number of fos- and Pdyn-co-expressing cells in the PVN and SON, but decreased it in the ARC. Deoxyglucose 7-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 263-266 32931071-7 2020 Afterward, the application of Baclofen and H89 alleviated the IS-evoked hyperalgesia and extenuated vesicular glutamate transporter 2 (VGLUT2), glutamate, calcitonin gene-related peptide (CGRP), and c-Fos expression by regulating the GABABR2/PKA/SynCAM1 pathway in the PAG of CM rats, while the application of CGP35348 and 8-Bromo-cAMP exactly exerted the opposite effect. Baclofen 30-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-204 32931071-7 2020 Afterward, the application of Baclofen and H89 alleviated the IS-evoked hyperalgesia and extenuated vesicular glutamate transporter 2 (VGLUT2), glutamate, calcitonin gene-related peptide (CGRP), and c-Fos expression by regulating the GABABR2/PKA/SynCAM1 pathway in the PAG of CM rats, while the application of CGP35348 and 8-Bromo-cAMP exactly exerted the opposite effect. N-(2-(4-bromocinnamylamino)ethyl)-5-isoquinolinesulfonamide 43-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-204 33375878-7 2020 Quantification of immunohistochemically labeled Fos + cells revealed that exposure to SMS in different age stages during the first postnatal month stimulates activity in cells of individual MOB/AOB layers in an age-dependent manner. sms 86-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 33375878-8 2020 In order to find out whether newly generated cells in the MOB/AOB could express Fos protein as a response to SMS, newborn rats were administrated with the marker of proliferation, bromodeoxyuridine (BrdU) at P0, and three weeks later (at P21) colocalization of Fos and BrdU in the neurons of the MOB and AOB was assessed. sms 109-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 33192203-0 2020 Boswellic Acid Synergizes With Low-Level Ionizing Radiation to Modulate Bisphenol Induced-Lung Toxicity in Rats by Inhibiting JNK/ERK/c-Fos Pathway. boswellic acid 0-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-139 33192203-8 2020 Moreover, BA and or/gamma-R ameliorated the lung inflammation via regulation of the JNK/ERK/c-Fos and Nrf2/ HO-1 signaling pathways. boswellic acid 10-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 32628919-8 2020 CCK-8S (25-fold and 51-fold, p = 0.025 and p = 0.001) and BN (31-fold and 59-fold, p = 0.007 and p = 0.001) at both doses increased the number of c-Fos positive neurons in the NTS. cholecystokinin 8 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-151 32628919-10 2020 Both doses of CCK-8S (24-fold and 48-fold p=0.011 and p=0.001) and bombesin (31-fold and 56-fold, p=0.002 and p=0.001) increased the number of activated Nnat neurons in the NTS (p=0.001) compared to the vehicle group, while in the DMV no significant increase of c-Fos activation was detected. cholecystokinin 8 14-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 262-267 32661781-0 2020 The Combination of Cholecystokinin and Stress Amplifies an Inhibition of Appetite, Gastric Emptying, and an Increase in c-Fos Expression in Neurons of the Hypothalamus and the Medulla Oblongata. Cholecystokinin 19-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 32841609-11 2020 Results showed that CaMKII had a role in anxiety-like behavior in the offspring of morphine-exposed parents, and changes in neuronal firing rate and c-fos level in the NAC might be involved in this process. Morphine 83-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 149-154 32730865-3 2020 We investigated the effect of 3 weeks of Flx treatment (15 mg/kg/day) on changes in neuronal activity, by mapping the number of c-Fos+ cells, in several brain subregions in adult male rats of control and following 3 weeks of chronic social isolation (CSIS), an animal model of depression. Fluoxetine 41-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-133 32434091-5 2020 Similar to T3, TDCPP and BDCPP also significantly upregulated c-fos and downregulated Tshbeta gene expression. tris(1,3-dichloro-2-propyl)phosphate 15-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 32434091-5 2020 Similar to T3, TDCPP and BDCPP also significantly upregulated c-fos and downregulated Tshbeta gene expression. bdcpp 25-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 32472169-5 2020 DON-induced neuronal activation was assessed by c-Fos immunohistochemistry. deoxynivalenol 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 32472169-6 2020 DON injection resulted in profound c-Fos activation in only the elements of the reward system, such as the accumbens nucleus, the medial prefrontal cortex, and the ventral tegmental area. deoxynivalenol 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 32673629-6 2020 Intra-ACC administration of CID16020046 prevented the formalin-induced increases in expression of mRNA coding for the immediate early gene and marker of neuronal activity, c-Fos, in the ipsilateral dorsal horn of the spinal cord. 4-(4-(3-hydroxyphenyl)-3-(4-methylphenyl)-6-oxo-1H,4H,5H,6H-pyrrolo(3,4-c)pyrazol-5-yl)benzoic acid 28-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-177 32673629-6 2020 Intra-ACC administration of CID16020046 prevented the formalin-induced increases in expression of mRNA coding for the immediate early gene and marker of neuronal activity, c-Fos, in the ipsilateral dorsal horn of the spinal cord. Formaldehyde 54-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-177 32914562-9 2020 Fos expression showed neuronal activation in the brain regions that are associated with the hypothalamus and/or are involved in maintaining water and electrolyte homeostasis in HTN- and PEG-treated rats. Water 140-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 32914562-9 2020 Fos expression showed neuronal activation in the brain regions that are associated with the hypothalamus and/or are involved in maintaining water and electrolyte homeostasis in HTN- and PEG-treated rats. Polyethylene Glycols 186-189 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 33062018-5 2020 The effects of TMP on the protein expression of FOS and acid sensing ion channel1a (ASIC1a) in the brainstem induced by hypoxia were investigated by immunohistochemistry. tetramethylpyrazine 15-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 33062018-7 2020 Hypoxia obviously induced the protein expression of FOS (P < 0.01) and ASIC1a(P < 0.05) in the brainstem, which can be also significantly inhibited by TMP pretreatment. tetramethylpyrazine 151-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 33062018-8 2020 Conclusions: TMP has protective effects on hypoxic respiratory depression, and the mechanisms might be concerned with its downregulation of FOS and ASIC1a in the brainstem induced by hypoxia. tetramethylpyrazine 13-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-143 32957694-7 2020 Immunohistochemical analyses revealed that minocycline inhibited the increase in c-Fos immune-reactive (IR) cells and the fluorescence intensity of both Iba1 and glial fibrillary acidic protein (GFAP) in the Vc following IONI. Minocycline 43-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 32957694-8 2020 Intracisternal administration of C1q caused orofacial mechanical hypersensitivity and an increase in the number of c-Fos-IR cells and fluorescence intensity of GFAP. C10Ph 33-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 32717296-9 2020 Significant increases in the numbers of c-Fos-positive neurons in the unpaired group suggest that LHb neurons engage in the process that associates a CS with the absence of an US. Cesium 150-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 32682656-4 2020 We measured both incentive motivation for cocaine using progressive ratio procedures, and cocaine-induced c-fos mRNA expression, a marker of neuronal activity. Cocaine 90-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 32682656-12 2020 Compared to LgA-90 s rats, IntA-5 s rats had more cocaine-induced c-fos mRNA in the orbitofrontal and prelimbic cortices and the caudate-putamen. Cocaine 50-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 32817784-6 2020 Confocal microscopy revealed that diazoxide also led to nuclear translocation of the transcription factors c-Fos and NFkappaB, which was also blocked by NAC or 5-HD. Diazoxide 34-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 33364051-10 2021 Additionally, D1DR/D2DR antagonists inhibited spinal Calcitonin Gene-Related Peptide (CGRP) and c-Fos expression and alleviated bone cancer pain induced by TCI which could both be reversed by NMDA. N-Methylaspartate 192-196 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 32848657-9 2020 The abolition of F-CPA following optogenetic inhibition of glutamatergic neurons of the ACC was associated with a reduction in c-Fos immunoreactivity in the ACC in male rats, but not female rats. f-cpa 17-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 32817784-6 2020 Confocal microscopy revealed that diazoxide also led to nuclear translocation of the transcription factors c-Fos and NFkappaB, which was also blocked by NAC or 5-HD. Acetylcysteine 153-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 32817784-8 2020 Conclusions: Our results suggest that opening mitochondrial potassium ATP channels with diazoxide upregulates the expression of STIM1 and Orai1 by de novo synthesis by a mechanism that involves NFkB, c-Fos, and ROS via MAPK/ERK signaling. Diazoxide 88-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 200-205 32512137-5 2020 Furthermore, corticosterone administration into male rats as a rodent model of depression induced significantly higher c-Fos expression in 5-HT3AR-positive GABAergic neurons compared to that in 5-HT neurons within the DRN. Corticosterone 13-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 32512137-5 2020 Furthermore, corticosterone administration into male rats as a rodent model of depression induced significantly higher c-Fos expression in 5-HT3AR-positive GABAergic neurons compared to that in 5-HT neurons within the DRN. Serotonin 139-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 32697679-6 2020 In Gq DREADD-injected rats, CNO induced significant increases in Fos staining in the MnPO and in regions that receive direct or indirect projections from the MnPO. gq dreadd 3-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 32376461-6 2020 Release of glutamate from nanoclusters in vivo caused enhanced c-Fos expression, indicating that the loading capacity of the nanoclusters is sufficient to induce neuronal activation. Glutamic Acid 11-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 31945187-7 2020 Furthermore, gene expression studies revealed that the promoters of AT-1, ATase1, and ATase2 contain functional binding sites for the neuron-related transcription factors cAMP response element-binding protein and the immediate-early genes c-FOS and c-JUN, and that ATase1 and ATase2 exhibit additional modes of transcriptional regulation relevant to aging and Alzheimer"s disease. Cyclic AMP 171-175 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 239-244 32627950-3 2020 METHODS: This study first explored whether iPBN cells respond to ghrelin involving Fos mapping and electrophysiological studies in rats. ipbn 43-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 32060380-5 2020 Furthermore, systemic PTX administration decreased c-Fos expression within the hypothalamic paraventricular nucleus (PTX: 17 +- 4 vs. Veh: 70 +- 13 cells, P < 0.01, N = 5, PVN) and increased the total number of microglial branches (PTX: 2129 +- 242 vs. Veh: 1415 +- 227 branches, P < 0.05, N = 4/group). Pentoxifylline 22-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 32699194-6 2020 Quantity of DCA-activated neurons in the hypothalamus was determined by c-Fos expression. Deoxycholic Acid 12-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 32699194-7 2020 Combining DCA and CCK-8 caused a 4-fold increase in c-Fos activation. Deoxycholic Acid 10-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 32699194-7 2020 Combining DCA and CCK-8 caused a 4-fold increase in c-Fos activation. Sincalide 18-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 32699194-8 2020 In the arcuate nucleus, c-Fos-positive neurons coexpressed cocaine and amphetamine regulated transcript and proopiomelanocortin. Cocaine 59-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 32699194-8 2020 In the arcuate nucleus, c-Fos-positive neurons coexpressed cocaine and amphetamine regulated transcript and proopiomelanocortin. Amphetamine 71-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 32699194-9 2020 DCA-induced c-Fos expression was eliminated following truncal vagotomy or silencing of TGR5 in the NG. Deoxycholic Acid 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 32353393-7 2020 RESULTS: Locomotor activity and c-Fos IR changes induced by THC challenge were altered by nicotine pre-exposure and modified by age and sex. Dronabinol 60-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 32353393-7 2020 RESULTS: Locomotor activity and c-Fos IR changes induced by THC challenge were altered by nicotine pre-exposure and modified by age and sex. Nicotine 90-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 32353393-10 2020 THC increased c-Fos IR in the caudate, nucleus accumbens, stria terminalis, septum, amygdala, hypothalamus, and thalamus. Dronabinol 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 32472163-10 2020 Moreover, Bexa blocked the sleep rebound period after total sleep deprivation, increased in the hypothalamus the expression of c-Fos, and decreased NeuN activity. Bexarotene 10-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 32472163-14 2020 Moreover, Bexa or UVI administration significantly affected c-Fos and NeuN expression in the hypothalamus. Bexarotene 10-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 32610694-7 2020 Additionally, the expression of phosphorylated (p) ERK, pCREB, c-Fos, and FosB/DeltaFosB in the NAc was significantly enhanced by 1.0 mg/kg 4"-F-PCP self-administration. 4"-f-pcp 140-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 32571909-5 2020 C-Fos experiments showed that the PSTN complex reacts to neophobia and sickness induced by lipopolysaccharide or cisplatin. Cisplatin 113-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 32571910-9 2020 Moreover, systemic administration of acetazolamide immediately after the extinction training session modulated c-Fos expression on a retention test in the ventromedial prefrontal cortex of rats trained in contextual fear conditioning. Acetazolamide 37-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 32802288-10 2020 Conclusion: The finding indicates that PBM treatment protects rat retina against light damage via the prevention of Fos, Ccl2, Icam1, Cxcl10, and Myc dysregulation. pbm 39-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 32248751-6 2020 Mitragynine-induced changes in c-fos expression were also analyzed. mitragynine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 32248751-7 2020 RESULTS: Mitragynine increased delta power and reduced theta power in all three cortical regions that were accompanied by increased c-fos expression. mitragynine 9-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 32555429-0 2020 Fatigue-induced Fos immunoreactivity within the lumbar cord and amygdala decreases after S60 fullerene pretreatment. s60 fullerene 89-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 32575871-10 2020 In parallel, 4-FS enhanced GABAA receptor expression, reduced ratio of glutamatergic GluN2A/2B receptors and enhanced the expression of Fos, a marker of neuronal activation in the ventral hippocampus in ED rats. 4-fs 13-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-139 32555429-4 2020 C60 fullerene pretreatment in animals with subsequent electrical stimulation induced a distinct (2-4 times) decrease in the level of Fos immunoreactivity in the observed structures in comparison with only fatigue-induced rats. Fullerenes 4-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-136 32597151-0 2020 Effect of amisulpride, olanzapine, quetiapine, and aripiprazole single administration on c-Fos expression in vasopressinergic and oxytocinergic neurons of the rat hypothalamic supraoptic nucleus. Amisulpride 10-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 32505192-9 2020 Subsequently, PMA induced MMP-9 expression via PKCdelta-mediated reactive oxygen species (ROS) generation, extracellular signal-regulated kinase 1/2 (ERK1/2) activation, and then induced c-Fos/AP-1 signaling pathway. Tetradecanoylphorbol Acetate 14-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 187-192 32224159-0 2020 Cyclosomatostatin-induced catalepsy in aged rats: Specific change of brain c-Fos protein expression in the lateral entorhinal cortex. cyclosomatostatin 0-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 31698029-6 2020 Ethanol significantly increased cFos expression in the MO region for control (p < 0.0001), NTX (p< 0.05), Aya1 (p<0.001) and Aya2 (p< 0.0001) groups. Ethanol 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-36 31698029-6 2020 Ethanol significantly increased cFos expression in the MO region for control (p < 0.0001), NTX (p< 0.05), Aya1 (p<0.001) and Aya2 (p< 0.0001) groups. Naltrexone 94-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-36 31698029-8 2020 Furthermore, NTX and Aya0.5 treatment decreased cFos expression compared to control in the MO region (p< 0.05 and p<0.01, respectively), but only the ayahuasca group reached levels not significantly different from the naive group. Naltrexone 13-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-52 32147623-7 2020 In the rat formalin test, oral administration of ETZ significantly reduced the nociceptive responses of the second phase and also the number of c-Fos-expressing cells in the spinal dorsal horn, in a dose-dependent manner. ethenzamide 49-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-149 32070769-7 2020 However, the neurite length, activity-regulated cytoskeleton-associated protein (Arc) and c-Fos mRNA were inhibited with FLU exposure at day 3. Fluoxetine 121-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 32482053-10 2020 Dexmedetomidine treatment alleviated thermal hyperalgesia and downregulated expression of c-Fos in the vlPAG and PVN after SNCI. Dexmedetomidine 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 32147481-11 2020 Additionally, EVO suppressed serum NO levels and reduced the mRNA/protein expression of c-Fos and nNOS, but not iNOS, in the PAG. evodiamine 14-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 32341122-6 2020 Daily oxycodone administration (2 mg/kg) increased pain thresholds and increased Fos+ neurons in the basolateral amygdala during intoxication, with a decrease in pain thresholds and increase in Fos+ neurons in the periaqueductal gray, central nucleus of the amygdala, locus coeruleus, paraventricular nucleus of the thalamus, agranular insular cortex, bed nucleus of the stria terminalis, and lateral habenula medial parvocellular region during withdrawal. Oxycodone 6-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-84 32341122-6 2020 Daily oxycodone administration (2 mg/kg) increased pain thresholds and increased Fos+ neurons in the basolateral amygdala during intoxication, with a decrease in pain thresholds and increase in Fos+ neurons in the periaqueductal gray, central nucleus of the amygdala, locus coeruleus, paraventricular nucleus of the thalamus, agranular insular cortex, bed nucleus of the stria terminalis, and lateral habenula medial parvocellular region during withdrawal. Oxycodone 6-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 194-197 32547170-12 2020 In phase 2 study, both NR2B subunit antagonist Ro25-6981 and iwp-2 decreased the amount of activated NR2B, enhanced p-GSK-3beta (Ser9), reduced beta-catenin, c-fos and NF-kappaB in the lumbar spinal cord (p < 0.001). Ro 25-6981 47-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 31630385-11 2020 The expression of c-Fos in C-fibers in the spinal cord (L6) decreased significantly after 1% lidocaine treatment in rats with BOO. Lidocaine 93-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 32429243-5 2020 PEAum-Paracetamol association was able to reduce hyperalgesia, mast cell activation, c-Fos and nerve growth factor (NGF) expression, neural histological damage, cytokine release, and apoptosis. peaum-paracetamol 0-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 32200030-7 2020 Early flupenthixol treatment was associated with more apomorphine-induced c-Fos labeling in the nucleus accumbens shell than the early saline-apomorphine group, indicating a sensitized response. Flupenthixol 6-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 32200030-7 2020 Early flupenthixol treatment was associated with more apomorphine-induced c-Fos labeling in the nucleus accumbens shell than the early saline-apomorphine group, indicating a sensitized response. Apomorphine 54-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 32597151-0 2020 Effect of amisulpride, olanzapine, quetiapine, and aripiprazole single administration on c-Fos expression in vasopressinergic and oxytocinergic neurons of the rat hypothalamic supraoptic nucleus. Aripiprazole 51-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 32597151-8 2020 With respect to the stimulation efficacy of the individual antipsychotics, estimated based on the quantity of c-Fos-labeled AVP and OXY neurons, could be a preferential action assigned to QUE over moderate effect of ARI and lower effect to OLA and reduced effect of AMI (VEH < AMI < OLA < ARI < QUE). Olanzapine 240-243 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 32597151-8 2020 With respect to the stimulation efficacy of the individual antipsychotics, estimated based on the quantity of c-Fos-labeled AVP and OXY neurons, could be a preferential action assigned to QUE over moderate effect of ARI and lower effect to OLA and reduced effect of AMI (VEH < AMI < OLA < ARI < QUE). Amisulpride 266-269 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 32235766-6 2020 In addition, both E2 and FLX augmented the c-Fos expression in the SCN, specifically during the light phase. Estradiol 18-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 32235506-0 2020 Antagonism of Histamine H3 receptors Alleviates Pentylenetetrazole-Induced Kindling and Associated Memory Deficits by Mitigating Oxidative Stress, Central Neurotransmitters, and c-Fos Protein Expression in Rats. Pentylenetetrazole 48-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 178-183 32235766-6 2020 In addition, both E2 and FLX augmented the c-Fos expression in the SCN, specifically during the light phase. flx 25-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 31571657-7 2020 Using dual immunolabeling for Fos protein, a marker for neuronal activity, and serotonin (5-hydroxytrypamine; 5-HT), numbers of Fos-positive 5-HT neurons were determined in five dorsal raphe nuclei. Serotonin 141-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-131 32074544-6 2020 Fluoxetine inhibited increases in the number of Fos-positive neurons in the NRM that occurred as a result of FSTs, but this was not observed in sham rats. Fluoxetine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 32051327-8 2020 Relapse to fentanyl seeking was associated with increased Fos expression in piriform cortex (Pir) neurons projecting to OFC, but not in projections from basolateral amygdala and thalamus. Fentanyl 11-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 31571657-8 2020 We found that restraint stress alone increased numbers of Fos-positive 5-HT neurons in all five dorsal raphe nuclei compared to control animals. Serotonin 71-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 31571657-9 2020 However, following P3 HI, the number of stress-induced Fos-positive 5-HT neurons was decreased significantly in the dorsal raphe ventrolateral, interfascicular and ventral nuclei compared with control animals exposed to restraint stress. Serotonin 68-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 31571657-10 2020 In contrast, numbers of stress-induced Fos-positive 5-HT neurons in the dorsal raphe dorsal and caudal nuclei were not affected by P3 HI. Serotonin 52-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 31066114-10 2020 We then examined inter-regional correlations in Fos-protein expression for all 42 brain regions, which showed Fos expression was more strongly positively correlated following yohimbine-induced reinstatement of alcohol seeking, compared to home-cage yohimbine. Alcohols 210-217 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 31066114-8 2020 The number of Fos-protein positive nuclei was increased in the prefrontal cortex and extended amygdala after home-cage yohimbine compared to naive- and vehicle-treated rats. Yohimbine 119-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 31066114-9 2020 Yohimbine-induced reinstatement increased the number of Fos-protein expressing nuclei in multiple other regions including the thalamus, hypothalamus and hippocampus. Yohimbine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 31711895-8 2020 Similarly, ERalpha-agonism (PPT) increased Fos and Homer1 induction ~3-fold in WT and LVR isolated mPOA neurons, with no effect of the ERbeta-agonist DPN alone or in combination with PPT, suggesting medial-preoptic ERbeta activity may blunt ERalpha signaling. 4,4',4''-(4-propyl-((1)H)-pyrazole-1,3,5-triyl) tris-phenol 28-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 31266052-9 2020 A history of cocaine + alcohol also altered patterns of reinstatement-induced Fos expression. Cocaine 13-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 32010247-11 2020 High glucose levels also upregulated the expression and activation of PKC-alpha, PKC-beta2, NF-kappaB, TNF-alpha and c-fos. Glucose 5-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 32010247-13 2020 In conclusion, metoprolol and bisoprolol could prevent hypertrophy of cardiomyocytes cultured in high glucose by the inhibition of the total and phospho-PKC-alpha, which could further influence the PKC-alpha/NF-kappaB/c-fos signaling pathway. Metoprolol 15-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 218-223 32010247-13 2020 In conclusion, metoprolol and bisoprolol could prevent hypertrophy of cardiomyocytes cultured in high glucose by the inhibition of the total and phospho-PKC-alpha, which could further influence the PKC-alpha/NF-kappaB/c-fos signaling pathway. Bisoprolol 30-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 218-223 31066114-10 2020 We then examined inter-regional correlations in Fos-protein expression for all 42 brain regions, which showed Fos expression was more strongly positively correlated following yohimbine-induced reinstatement of alcohol seeking, compared to home-cage yohimbine. Yohimbine 175-184 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 31066114-10 2020 We then examined inter-regional correlations in Fos-protein expression for all 42 brain regions, which showed Fos expression was more strongly positively correlated following yohimbine-induced reinstatement of alcohol seeking, compared to home-cage yohimbine. Yohimbine 175-184 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 32010247-0 2020 Metoprolol and bisoprolol ameliorate hypertrophy of neonatal rat cardiomyocytes induced by high glucose via the PKC/NF-kappaB/c-fos signaling pathway. Metoprolol 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 32010247-0 2020 Metoprolol and bisoprolol ameliorate hypertrophy of neonatal rat cardiomyocytes induced by high glucose via the PKC/NF-kappaB/c-fos signaling pathway. Bisoprolol 15-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 32010247-0 2020 Metoprolol and bisoprolol ameliorate hypertrophy of neonatal rat cardiomyocytes induced by high glucose via the PKC/NF-kappaB/c-fos signaling pathway. Glucose 96-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 31629892-2 2020 We previously reported that the peripherally restricted FAAH inhibitor URB937, which selectively increases AEA levels outside the central nervous system, reduces hyperalgesia and c-Fos expression in the trigeminal nucleus caudalis (TNC) and the locus coeruleus in an animal model of migraine based on nitroglycerin (NTG) administration. URB937 71-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-184 31629892-6 2020 RESULTS: Pre-treatment with URB597 significantly reduced c-Fos immunoreactivity in the TNC and inhibited NTG-induced hyperalgesia in the orofacial formalin test. cyclohexyl carbamic acid 3'-carbamoylbiphenyl-3-yl ester 28-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 31392556-11 2020 Renal infusion of capsaicin increased c-Fos expression in the paraventricular nucleus (PVN) of hypothalamus. Capsaicin 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 31778691-7 2020 We found that intra-LHb infusion of SB242084, a selective 5-HT2CR antagonist alleviated AB and reduced the elevated c-Fos expression in the LHb of Post-EtOH rats. 6-chloro-5-methyl-1-((2-(2-methylpyrid-3-yloxy)pyrid-5-yl)carbamoyl)indoline 36-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 31778691-8 2020 By contrast, intra-LHb infusion of the selective 5-HT2CR agonist WAY161503 induced AB and increased c-Fos expression in the LHb in alcohol-naive but not Post-EtOH rats. 5-[[[(2~{r},3~{s},4~{r},5~{r})-5-(6-Aminopurin-9-Yl)-3,4-Bis(Oxidanyl)oxolan-2-Yl]methylamino]methyl]-4-Azanyl-1-(Methoxymethyl)pyrimidin-2-One 19-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 31778691-8 2020 By contrast, intra-LHb infusion of the selective 5-HT2CR agonist WAY161503 induced AB and increased c-Fos expression in the LHb in alcohol-naive but not Post-EtOH rats. 8,9-dichloro-2,3,4,4a-tetrahydro-1H-pyrazino(1,2-a)quinoxalin-5(6H)-one 65-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 31934719-13 2020 Emodin significantly abolished IBD-enhanced Traf6, NFATC1 and c-fos expression. Emodin 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 32009899-1 2019 To clarify the different regional brain electroencephalogram (EEG) activities and biochemical responses in seizure and epilepsy models, we assessed the EEG and c-Fos immunolabeling characteristics in a lithium-pilocarpine-induced status epilepticus (SE) model and pentylenetetrazol (PTZ)-induced seizure model. Pilocarpine 202-221 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 32009899-7 2019 The labeling of c-Fos was enhanced in the cortex and hippocampal CA1, CA3, and dentate gyrus (DG); however, compared to the PTZ-induced seizure model, c-Fos staining could only be observed in the striatum and thalamus in the lithium-pilocarpine-induced epilepsy model. Pentylenetetrazole 124-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 32009899-7 2019 The labeling of c-Fos was enhanced in the cortex and hippocampal CA1, CA3, and dentate gyrus (DG); however, compared to the PTZ-induced seizure model, c-Fos staining could only be observed in the striatum and thalamus in the lithium-pilocarpine-induced epilepsy model. Pilocarpine 225-244 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 32009899-8 2019 In each brain region, prominent c-Fos labeling was observed 2 h and 4 h after the induction of SE or seizures and diminished at 24 h. During the lithium-pilocarpine-induced chronic epilepsy phase after SE induction, MFS was observed 7 days after SE and was accompanied by the dynamic evolution of epileptic EEG activities. Pilocarpine 145-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 32009899-10 2019 The EEG characteristics and c-Fos expression patterns differ from those presented in a previous study using a PTZ-induced seizure model. Pentylenetetrazole 110-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 31266052-9 2020 A history of cocaine + alcohol also altered patterns of reinstatement-induced Fos expression. Alcohols 23-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 31704349-0 2020 c-Fos Expression after Stochastic Vestibular Stimulation and Levodopa in 6-OHDA Hemilesioned Rats. Oxidopamine 73-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 32021391-8 2020 Bilateral block with a relatively small volume of bupivacaine (0.5 mL) significantly increased the threshold to mechanical pain as compared to control (p=0.007) and significantly decreased the number of c-Fos immunoreactive nuclei in the posterior horn of the spinal cord (p<0.0001). Bupivacaine 50-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 203-208 31704349-7 2020 SVS and levodopa induced similar c-Fos expression in several regions, e.g. the caudate putamen (CPu), where saline had no effect. Levodopa 8-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 31722249-8 2020 Histopathological changes and immunohistochemical expressions of c-Jun N-terminal kinase (JNK) and c-Fos in the IFA-induced brain tissues were decreased after administration of morin. Ifosfamide 112-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 31926296-6 2020 The highest density of c-Fos-ir cells (c-Fos - a marker of neuronal activation) was demonstrated by the medial nucleus (Me) and bed nucleus of the accessory olfactory tract (BAOT). (5-amino-1,3-dimethylpyrazol-4-yl)-(2-fluorophenyl)methanone 119-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 31926296-6 2020 The highest density of c-Fos-ir cells (c-Fos - a marker of neuronal activation) was demonstrated by the medial nucleus (Me) and bed nucleus of the accessory olfactory tract (BAOT). (5-amino-1,3-dimethylpyrazol-4-yl)-(2-fluorophenyl)methanone 119-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 32785417-10 2020 Results A statistically significant increase (p<0.01) in c-Fos+ cells was noted in nephropathic animals receiving star fruit juice compared to control groups, in brain areas commonly related to epileptogenic neural circuits including the hippocampus, amygdala, rhinal cortex, anterior cingulate area, piriform area, and medial dorsal thalamus. star fruit juice 114-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 31704349-6 2020 Furthermore, c-Fos expression increased more in the habenula nucleus (LHb) after SVS than it did after levodopa in 6-OHDA hemilesioned animals and after saline in the sham-lesioned animals. Levodopa 103-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 31704349-6 2020 Furthermore, c-Fos expression increased more in the habenula nucleus (LHb) after SVS than it did after levodopa in 6-OHDA hemilesioned animals and after saline in the sham-lesioned animals. Oxidopamine 115-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 31852437-7 2019 To evaluate the developmentally generated granule neurons that support hippocampus-dependent memory, spatial reference memory was tested by using Morris Water Maze at 3 months old, after which the immunofluorescence was used to detect c-Fos expression in the NeuN+/BrdU+ cells. neuronal nuclear antigen NeuN, human 259-263 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 235-240 31852437-7 2019 To evaluate the developmentally generated granule neurons that support hippocampus-dependent memory, spatial reference memory was tested by using Morris Water Maze at 3 months old, after which the immunofluorescence was used to detect c-Fos expression in the NeuN+/BrdU+ cells. Bromodeoxyuridine 265-269 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 235-240 31678081-0 2019 Fos expression induced by olanzapine and risperidone in the central extended amygdala. Olanzapine 26-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 31678081-0 2019 Fos expression induced by olanzapine and risperidone in the central extended amygdala. Risperidone 41-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 31678081-4 2019 Fos-like-immunoreactivity induced by olanzapine and risperidone was compared with that by the atypical antipsychotic clozapine and typical antipsychotic haloperidol. Olanzapine 37-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 31678081-4 2019 Fos-like-immunoreactivity induced by olanzapine and risperidone was compared with that by the atypical antipsychotic clozapine and typical antipsychotic haloperidol. Risperidone 52-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 31678081-5 2019 Regarding the extended amygdala, and similarly to clozapine, olanzapine (5-10 mg/kg) and, with a lower efficacy, risperidone (1-3 mg/kg), induced Fos-like-nuclei in CeA, IPAC, SLEA, and BSTL. Olanzapine 61-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-149 31678081-5 2019 Regarding the extended amygdala, and similarly to clozapine, olanzapine (5-10 mg/kg) and, with a lower efficacy, risperidone (1-3 mg/kg), induced Fos-like-nuclei in CeA, IPAC, SLEA, and BSTL. Risperidone 113-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-149 31678081-7 2019 On the contrary, the increase of Fos-like-nuclei in the extended amygdala by haloperidol was restricted to IPAC only. Haloperidol 77-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 31678081-8 2019 These findings, consistent with the important role of extended amygdala in neuropsychiatric disorders characterized by affective disturbances, showed that olanzapine and risperidone, contrary to haloperidol, preferentially activated Fos-expression in these brain areas. Olanzapine 155-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 233-236 31678081-8 2019 These findings, consistent with the important role of extended amygdala in neuropsychiatric disorders characterized by affective disturbances, showed that olanzapine and risperidone, contrary to haloperidol, preferentially activated Fos-expression in these brain areas. Risperidone 170-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 233-236 31712057-4 2019 Female ovariectomized rats were injected with 17beta-estradiol (E2) or progesterone (P4) for 21 days, after which visceral pain-induced spinal c-fos expression and visceromotor reflex changes were compared between ovariectomized and hormone-substituted groups. Progesterone 71-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-148 31712057-5 2019 We found that uterine cervical distension induced a drastic increase in spinal c-fos expression and visceromotor reflex activity, and ovariectomy inhibited the increase in c-fos expression induced by visceral pain; this inhibition was reversed by estrogen but not progesterone replacement. Estrogens 247-255 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-177 31712057-5 2019 We found that uterine cervical distension induced a drastic increase in spinal c-fos expression and visceromotor reflex activity, and ovariectomy inhibited the increase in c-fos expression induced by visceral pain; this inhibition was reversed by estrogen but not progesterone replacement. Progesterone 264-276 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-177 30407216-3 2019 1,2-dimethylhydrazine-administered rats supplemented with p-CA showed downregulation of the expression of colonic proteins, namely, cyclin B1, cdc2, and mdm2, which regulate cell cycle, and immediate early response genes, namely, c-fos, c-jun and c-myc, which regulate cell proliferation. 1,2-Dimethylhydrazine 0-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 230-235 31805115-7 2019 Significant increase of c-Fos-IR cells in the Vc was found in 6-OHDA-injected rats after formalin administration compared with those in saline-injected rats after formalin administration. Oxidopamine 62-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 31805115-7 2019 Significant increase of c-Fos-IR cells in the Vc was found in 6-OHDA-injected rats after formalin administration compared with those in saline-injected rats after formalin administration. Formaldehyde 89-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 31805115-7 2019 Significant increase of c-Fos-IR cells in the Vc was found in 6-OHDA-injected rats after formalin administration compared with those in saline-injected rats after formalin administration. Formaldehyde 163-171 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 31805115-8 2019 We also assessed expression of c-Fos-IR cells in the paraventricular nucleus (PVN), and significant decrease of c-Fos-IR cells in the PVN of 6-OHDA-injected rats was found. Oxidopamine 141-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 31805115-8 2019 We also assessed expression of c-Fos-IR cells in the paraventricular nucleus (PVN), and significant decrease of c-Fos-IR cells in the PVN of 6-OHDA-injected rats was found. Oxidopamine 141-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 33223040-13 2020 Moreover, expressions of c-fos and c-jun genes that are prominent at 5h in untreated SCI are also considerably reduced by CBL and NWCBL treatment. Chlorambucil 122-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 33223040-13 2020 Moreover, expressions of c-fos and c-jun genes that are prominent at 5h in untreated SCI are also considerably reduced by CBL and NWCBL treatment. nwcbl 130-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 31511975-9 2019 When the neuroendocrine response to stress was explored, intra-dlPAG infusion of muscimol prior to stress decreased Fos expression in the paraventricular nucleus and serum corticosterone levels. dlpag 63-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 31511975-9 2019 When the neuroendocrine response to stress was explored, intra-dlPAG infusion of muscimol prior to stress decreased Fos expression in the paraventricular nucleus and serum corticosterone levels. Muscimol 81-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 31165503-3 2019 Recent work using c-fos expression as a metabolic marker for neural activity combined with temporary inactivation of relevant corticostriatal regions and pharmacological manipulations of opioid, cannabinoid, and dopamine systems has led to a better understanding of how basal ganglia circuitry may be involved in modulating social play in the juvenile rat. Cannabinoids 195-206 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 31165503-3 2019 Recent work using c-fos expression as a metabolic marker for neural activity combined with temporary inactivation of relevant corticostriatal regions and pharmacological manipulations of opioid, cannabinoid, and dopamine systems has led to a better understanding of how basal ganglia circuitry may be involved in modulating social play in the juvenile rat. Dopamine 212-220 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 30685495-9 2019 To confirm pain-dependent Mn enhancement in the spinal cord, c-Fos expression was analyzed, and was found to be increased in the formalin-injected rats. Formaldehyde 129-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 31539618-8 2019 In all regions of SSCx, Rt, VB, CM and LGN, GAERS caffeine group had lower c-Fos protein expression comparing to the GAERS control group (p < 0.05). Caffeine 50-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 31539618-9 2019 Wistar caffeine rats had lower expression of c-Fos protein comparing to the Wistar control group only in SSCx. Caffeine 7-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 30407216-3 2019 1,2-dimethylhydrazine-administered rats supplemented with p-CA showed downregulation of the expression of colonic proteins, namely, cyclin B1, cdc2, and mdm2, which regulate cell cycle, and immediate early response genes, namely, c-fos, c-jun and c-myc, which regulate cell proliferation. p-coumaric acid 58-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 230-235 31602208-11 2019 Puerarin upregulated peroxisome proliferator-activated receptor alpha (PPARalpha) and its downstream target genes nuclear respiratory factor 1, FOS proto-oncogene, YY1 transcription factor, acetyl-coenzyme A carboxylase a, Fas cell surface death receptor, L-type pyruvate kinase and acetyl-coenzyme A dehydrogenase medium chain in myocardial cells from rats with chronic heart failure. puerarin 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-147 31556948-9 2019 RESULTS: Prazosin blocked U50,488-induced reinstatement and decreased U50,488-induced Fos expression in the orbitofrontal cortex, nucleus accumbens core, ventral bed nucleus of the stria terminalis, central and basolateral amygdalar nuclei and ventral tegmental area. Prazosin 9-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 31527186-8 2019 SPS also altered c-Fos expression during fear and extinction learning/memory in midline thalamic nuclei. Sodium phenolsulfonate 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 31539618-11 2019 In conclusion differential effects of caffeine in the seizure modulation may involve c-Fos protein activity-dependent protection mechanisms. Caffeine 38-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 31802933-9 2019 The attenuation of c-fos immuno-positive cells in the ipsilateral trigeminal subnucleus caudalis after the intraperitoneal co-administration of ibuprofen (5 mg/kg) with dexamethasone (1 mg/kg) confirmed these synergistic antinociceptive effects. Ibuprofen 144-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 31802933-9 2019 The attenuation of c-fos immuno-positive cells in the ipsilateral trigeminal subnucleus caudalis after the intraperitoneal co-administration of ibuprofen (5 mg/kg) with dexamethasone (1 mg/kg) confirmed these synergistic antinociceptive effects. Dexamethasone 169-182 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 31686764-11 2019 RESULTS: TNBS treatment significantly increased the number of Fos-expressing neurons within the caudal NTS. Trinitrobenzenes 9-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 31058522-0 2019 Neuronal activation in orbitofrontal cortex subregions: Cfos expression following cue-induced reinstatement of cocaine-seeking behavior. Cocaine 111-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-60 31299320-0 2019 Clozapine increased c-Fos protein expression in several brain subregions of socially isolated rats. Clozapine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 31299320-2 2019 Here, we examined the effect of the atypical antipsychotic Clozapine (Clz) (20 mg/kg/day for 3 weeks) on the neuronal activation pattern of c-Fos protein expression in stress-relevant brain subregions of adult male Wistar rats exposed to chronic social isolation (CSIS: 3 weeks), an animal model of depression and schizophrenia, and controls. Clozapine 59-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 31299320-2 2019 Here, we examined the effect of the atypical antipsychotic Clozapine (Clz) (20 mg/kg/day for 3 weeks) on the neuronal activation pattern of c-Fos protein expression in stress-relevant brain subregions of adult male Wistar rats exposed to chronic social isolation (CSIS: 3 weeks), an animal model of depression and schizophrenia, and controls. Clozapine 70-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 31299320-5 2019 Increases in c-Fos protein expression in the RSGc, RSD, DGd, PVP, LA/BL complex of amygdala and striatum (CPu, Acb Core (AcbC) and AcbSh) following Clz treatment in controls were found. Clozapine 148-151 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 31299320-6 2019 Clz applied simultaneously with CSIS modulated neuronal activity in CPu, AcbC and AcbSh subregions compared to CSIS alone, increasing c-Fos protein expression. Clozapine 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-139 31542898-0 2019 Effect of lutein on the acute inflammation-induced c-Fos expression of rat trigeminal spinal nucleus caudalis and C1 dorsal horn neurons. Lutein 10-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 31542898-2 2019 The aim of the present study was to investigate whether pretreatment with lutein attenuates acute inflammation-induced sensitization of nociceptive processing in rat spinal trigeminal nucleus caudalis (SpVc) and upper cervical (C1) dorsal horn neurons, via c-Fos immunoreactivity. Lutein 74-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 257-262 31185197-8 2019 Furthermore, serotonin depletion also prevented the TBI-induced bilateral increase in c-Fos positive cells within the Rexed laminae I and II of the dorsal horns. Serotonin 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 31185197-8 2019 Furthermore, serotonin depletion also prevented the TBI-induced bilateral increase in c-Fos positive cells within the Rexed laminae I and II of the dorsal horns. Thioacetazone 52-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 31060041-6 2019 Using a chronic ethanol vapor exposure paradigm that renders rats physically dependent on ethanol, we observed significant withdrawal-induced enhancement of cFos expression in the RMTg. Ethanol 16-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-161 31060041-6 2019 Using a chronic ethanol vapor exposure paradigm that renders rats physically dependent on ethanol, we observed significant withdrawal-induced enhancement of cFos expression in the RMTg. Ethanol 90-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-161 31060041-6 2019 Using a chronic ethanol vapor exposure paradigm that renders rats physically dependent on ethanol, we observed significant withdrawal-induced enhancement of cFos expression in the RMTg. rmtg 180-184 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-161 31715034-8 2019 The results show that the expression of Fos in both vasopressin and oxytocin neurons of the PVN and SON were significantly elevated as soon as 90 min post-MI, compared to sham rats. Oxytocin 68-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 31715034-9 2019 Moreover, Fos protein was also elevated in tyrosine hydroxylase neurons in the nucleus tractus solitarius and rostral ventrolateral medulla of MI rats than sham rats. Tyrosine 43-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-13 31638891-4 2019 RESULTS: Chronic treatment with acetaminophen increased the frequency of cortical spreading depression (CSD) and the number of c-Fos-immunoreactive (Fos-IR) neurons in the trigeminal nucleus caudalis (TNC). Acetaminophen 32-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 31638891-4 2019 RESULTS: Chronic treatment with acetaminophen increased the frequency of cortical spreading depression (CSD) and the number of c-Fos-immunoreactive (Fos-IR) neurons in the trigeminal nucleus caudalis (TNC). Acetaminophen 32-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-132 31638891-5 2019 In the control group, muscimol microinjected into the NRM increased significantly the frequency of CSD-evoked direct current shift and Fos-IR neurons in the TNC. Muscimol 22-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-138 31638891-7 2019 In a model of migraine induced by intravenous systemic infusion of nitroglycerin (NTG), rats with chronic exposure to acetaminophen exhibited significantly more frequent neuronal firing in the TNC and greater Fos-IR than those without the acetaminophen treatment. Nitroglycerin 67-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 209-212 31638891-7 2019 In a model of migraine induced by intravenous systemic infusion of nitroglycerin (NTG), rats with chronic exposure to acetaminophen exhibited significantly more frequent neuronal firing in the TNC and greater Fos-IR than those without the acetaminophen treatment. Nitroglycerin 82-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 209-212 31638891-7 2019 In a model of migraine induced by intravenous systemic infusion of nitroglycerin (NTG), rats with chronic exposure to acetaminophen exhibited significantly more frequent neuronal firing in the TNC and greater Fos-IR than those without the acetaminophen treatment. Acetaminophen 118-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 209-212 31167109-10 2019 Running+memantine treatment increased adult neurogenesis by only 17%, but completely blocked experience-dependent Fos expression. Memantine 8-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 31374323-0 2019 Blunted hyperphagic and c-Fos immunoreactivity responsiveness to an orexigen, butorphanol tartrate, in aged rats. Butorphanol 78-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 31279580-0 2019 Spatial exploration induced expression of immediate early genes Fos and Zif268 in adult-born neurons Is reduced after pentylenetetrazole kindling. Pentylenetetrazole 118-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 31279580-9 2019 Using this approach, we found that PTZ kindled rats did not differ from control rats in regards to exploratory behaviour, but there was a marked attenuation in behaviour-induced expression of Fos and Zif268 for rats that received 2 weeks of PTZ kindling. Pentylenetetrazole 35-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 192-195 31279580-9 2019 Using this approach, we found that PTZ kindled rats did not differ from control rats in regards to exploratory behaviour, but there was a marked attenuation in behaviour-induced expression of Fos and Zif268 for rats that received 2 weeks of PTZ kindling. Pentylenetetrazole 241-244 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 192-195 31279580-10 2019 Further examination revealed that PTZ kindled rats showed reduced colocalization of Fos and Zif268 in 2.5 week old BrdU + cells. Pentylenetetrazole 34-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 31194948-9 2019 CCI attenuated restraint-induced Fos expression in LC neurons. CCI 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 31331999-8 2019 Here, we used the Daun02 chemogenetic inactivation procedure, which allows selective inhibition of neuronal ensembles identified by the activity marker Fos, to demonstrate that different ensembles for cocaine self-administration and extinction memories coexist in the ventral mPFC and interact with distinct subregions of the nucleus accumbens. Cocaine 201-208 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-155 31364821-0 2019 Distinct gene alterations between Fos-expressing striatal and thalamic neurons after withdrawal from methamphetamine self-administration. Methamphetamine 101-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 31557202-5 2019 The list of genes whose expression was affected by 1 mM ouabain by more than 2-fold was abundant in intermediates of intracellular signaling and transcription regulators, including augmented content of Npas4, Fos, Junb, Atf3, and Klf4 mRNAs, whose upregulated expression was demonstrated in neurons subjected to electrical and glutamatergic stimulation. Ouabain 56-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 209-212 31177064-9 2019 Additionally, ICA treatment significantly increased the neurogranin (Ng) and c-fos protein expression of hippocampus in the offspring rats. icariin 14-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 31177064-11 2019 CONCLUSION: Taken together, ICA may be an effective therapeutic for learning and memory dysfunction in female offspring exposed to PS, its neuroprotective effect was mediated in part by normalizing the HPA axis and up-regulating of ERK/CaMKIIalpha/CREB signaling, Ng and c-fos protein. icariin 28-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 271-276 31364821-3 2019 Here, we examined molecular alterations in DMS and AIT neurons activated (identified by neuronal activity marker Fos) during "incubated" Meth-seeking relapse test after prolonged withdrawal. Methamphetamine 137-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-116 30955244-16 2019 The mRNA levels of c-Fos, CGRP, GFAP, iba-1, and pro-inflammatory cytokines, also including Connexin 30 and Connexin 43 were decreased after injection of AMD3100, while EAAT 1 and EAAT 2 mRNAs were increased. plerixafor 154-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 31390064-4 2019 We observed an increased number of c-Fos-positive neurons in the ACC after injection of hypertonic saline, indicating strong ACC neuronal activation under hyperosmotic thirst. Sodium Chloride 99-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 31034840-12 2019 Quantification by in situ hybridization of c-Fos mRNA expression in the NTS 90 min post-gavage, showed a significant increase with each macronutrient, but was 24-29% higher with a lipid or peptone gavage compared to a glucose gavage. Glucose 218-225 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 31185278-10 2019 EtOH-PHY treatment rescued mPFC expression of c-Fos in ethanol-exposed rats and increased Arc and Npas4 regardless of dosing condition. Ethanol 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 31185278-10 2019 EtOH-PHY treatment rescued mPFC expression of c-Fos in ethanol-exposed rats and increased Arc and Npas4 regardless of dosing condition. Physostigmine 5-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 31185278-10 2019 EtOH-PHY treatment rescued mPFC expression of c-Fos in ethanol-exposed rats and increased Arc and Npas4 regardless of dosing condition. Ethanol 55-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 31201928-12 2019 Notably, in ORC but not in intact males, MK801 impairs CFM retrieval and expression of c-Fos and Egr-1 proteins in A30, without affecting their expression in limbic structures. Dizocilpine Maleate 41-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 31299348-9 2019 Evaluation of c-Fos expression as a marker of neural activation revealed that xanomeline increased the number of c-Fos-positive cells in several cortical areas, the hippocampal formation, amygdala, and nucleus accumbens. xanomeline 78-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 31299348-9 2019 Evaluation of c-Fos expression as a marker of neural activation revealed that xanomeline increased the number of c-Fos-positive cells in several cortical areas, the hippocampal formation, amygdala, and nucleus accumbens. xanomeline 78-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 31299348-10 2019 Other than in the orbital cortex and claustrum, TAK-071 induced similar c-Fos expression patterns. TAK-071 48-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 31299348-11 2019 When donepezil was co-administered to increase the levels of acetylcholine, the number of TAK-071-induced c-Fos-positive cells in these brain regions was increased. Donepezil 5-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 31299348-11 2019 When donepezil was co-administered to increase the levels of acetylcholine, the number of TAK-071-induced c-Fos-positive cells in these brain regions was increased. Acetylcholine 61-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 31299348-11 2019 When donepezil was co-administered to increase the levels of acetylcholine, the number of TAK-071-induced c-Fos-positive cells in these brain regions was increased. TAK-071 90-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 31185235-12 2019 Notably, allicin ameliorated the expression of KGF, Gadd45a, c-Fos, Dusp5 and Phospholipase A2, which were related to liver injury. allicin 9-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 31022457-7 2019 In addition, HFD exposure altered avoidance- or escape task-induced FOS-immunoreactivity within brain structures involved in control of neuroendocrine, autonomic, and behavioral responses to aversive stimuli, including the basolateral amygdala (BLA) and dorsomedial (DMH), paraventricular (PVN) and ventromedial (VMH) hypothalamic nuclei. Dimenhydrinate 267-270 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 31399090-8 2019 RESULTS: CR extract suppressed osteoclastogenesis, its activity and bone resorption activity through decreasing gene of osteoclast-related such as nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1), c-Fos, etc. Chromium 9-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 208-213 31447629-8 2019 Furthermore, TP, administered with or without FAD, induced the activation of Fos-immunoreactivity in brain areas implicated in sexual motivation and reward including the medial preoptic area, ventrolateral division of the ventromedial nucleus of the hypothalamus, the nucleus accumbens core, and the prefrontal cortex. Testosterone Propionate 13-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-80 30797833-0 2019 Fos activation patterns related to acute ethanol and conditioned taste aversion in adolescent and adult rats. Ethanol 41-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 30797833-4 2019 An adolescent-specific ethanol-induced increase in Fos+ staining was seen within the nucleus accumbens shell and core. Ethanol 23-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 30797833-5 2019 An age difference was also noted within the Edinger-Westphal nucleus (EW) following administration of the lower dose of ethanol, with 1 g/kg ethanol producing CTA in adults but not in adolescents and inducing a greater EW Fos response in adults than adolescents. Ethanol 120-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 222-225 30797833-5 2019 An age difference was also noted within the Edinger-Westphal nucleus (EW) following administration of the lower dose of ethanol, with 1 g/kg ethanol producing CTA in adults but not in adolescents and inducing a greater EW Fos response in adults than adolescents. Ethanol 141-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 222-225 30797833-8 2019 In the BNST (but not PrL), ethanol-induced increases in Fos-immunoreactivity (IR) were evident at both ages. Ethanol 27-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 30916039-11 2019 Knockdown of ROR2 in the spinal cord prevented and reversed CCI-induced pain behaviours and spinal neuronal sensitisation [Fos expression: 130 (12) vs 81 (8) cells, P<0.05; 120 (11) vs 70 (7) cells, P<0.05]. CCI 60-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-126 31317263-0 2019 Isoflurane induces c-Fos expression in the area postrema of the rat. Isoflurane 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 31317263-3 2019 In this study, we assessed whether isoflurane induced the expression of c-Fos, a neuronal activation marker, in the area postrema (AP), the locus of the CTZ, in rats, which do not have vomiting action. Isoflurane 35-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 31317263-7 2019 RESULTS: One-way analysis of variance showed that isoflurane exposure significantly increased c-Fos expression in the AP; however, the rats pretreated with 4 mg/kg ondansetron showed significantly decreased c-Fos expression. Isoflurane 50-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 31317263-7 2019 RESULTS: One-way analysis of variance showed that isoflurane exposure significantly increased c-Fos expression in the AP; however, the rats pretreated with 4 mg/kg ondansetron showed significantly decreased c-Fos expression. Ondansetron 164-175 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 31317263-7 2019 RESULTS: One-way analysis of variance showed that isoflurane exposure significantly increased c-Fos expression in the AP; however, the rats pretreated with 4 mg/kg ondansetron showed significantly decreased c-Fos expression. Ondansetron 164-175 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 207-212 31005664-5 2019 Intrathecal administration of RS504393 (a CCR2 antagonist) markedly reduced the zymosan-induced thermal and mechanical hyperalgesia accompanied with reduced expression in the spinal cord of c-Fos, CD11b, phosphorylated p38 mitogen activated protein kinases (p-p38), and interleukin-1beta. RS 504393 30-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 190-195 31119435-6 2019 The SMGI, astrocyte and neuron activity were affected after the astrocytotoxin L-A-aminohexanedioic acid (L-AA) and c-fos antisense oligonucleotide (ASO) injections. Oligonucleotides 132-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 31119435-6 2019 The SMGI, astrocyte and neuron activity were affected after the astrocytotoxin L-A-aminohexanedioic acid (L-AA) and c-fos antisense oligonucleotide (ASO) injections. Oligonucleotides, Antisense 149-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 30930149-7 2019 Moreover, phoenixin injected intracerebroventricularly (icv) activated several nuclei throughout the rat brain as assessed using c-Fos; the number of c-Fos/nesfatin-1 immunoreactive neurons was increased in the lateral septal nucleus (4-fold), supraoptic nucleus (107-fold), paraventricular nucleus (6-fold) and the nucleus of the solitary tract (18-fold) compared to vehicle (p < 0.05). phoenixin 10-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 30930149-7 2019 Moreover, phoenixin injected intracerebroventricularly (icv) activated several nuclei throughout the rat brain as assessed using c-Fos; the number of c-Fos/nesfatin-1 immunoreactive neurons was increased in the lateral septal nucleus (4-fold), supraoptic nucleus (107-fold), paraventricular nucleus (6-fold) and the nucleus of the solitary tract (18-fold) compared to vehicle (p < 0.05). phoenixin 10-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-155 31517634-0 2019 Spatial relationship between the c-Fos distribution and enkephalinergic, substance P, and tyrosine hydroxylase innervation fields after acute treatment with neuroleptics olanzapine, amisulpride, quetiapine, and aripiprazole in the rat septum. Olanzapine 170-180 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 31517634-0 2019 Spatial relationship between the c-Fos distribution and enkephalinergic, substance P, and tyrosine hydroxylase innervation fields after acute treatment with neuroleptics olanzapine, amisulpride, quetiapine, and aripiprazole in the rat septum. Amisulpride 182-193 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 31517634-0 2019 Spatial relationship between the c-Fos distribution and enkephalinergic, substance P, and tyrosine hydroxylase innervation fields after acute treatment with neuroleptics olanzapine, amisulpride, quetiapine, and aripiprazole in the rat septum. Aripiprazole 211-223 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 31517634-1 2019 OBJECTIVE: The aim of the present study was to demonstrate the spatial relationship between the c-Fos immunoreactive cells elicited by an acute treatment with neuroleptics including amisulpride (AMI), olanzapine (OLA), quetiapine (QUE), and aripiprazole (ARI) and enkephalinergic (ENK), substance P (SP), and tyrosine hydroxylase (TH) innervation fields in the rat septum. Amisulpride 182-193 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 31517634-1 2019 OBJECTIVE: The aim of the present study was to demonstrate the spatial relationship between the c-Fos immunoreactive cells elicited by an acute treatment with neuroleptics including amisulpride (AMI), olanzapine (OLA), quetiapine (QUE), and aripiprazole (ARI) and enkephalinergic (ENK), substance P (SP), and tyrosine hydroxylase (TH) innervation fields in the rat septum. Amisulpride 195-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 31517634-1 2019 OBJECTIVE: The aim of the present study was to demonstrate the spatial relationship between the c-Fos immunoreactive cells elicited by an acute treatment with neuroleptics including amisulpride (AMI), olanzapine (OLA), quetiapine (QUE), and aripiprazole (ARI) and enkephalinergic (ENK), substance P (SP), and tyrosine hydroxylase (TH) innervation fields in the rat septum. Olanzapine 201-211 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 31517634-1 2019 OBJECTIVE: The aim of the present study was to demonstrate the spatial relationship between the c-Fos immunoreactive cells elicited by an acute treatment with neuroleptics including amisulpride (AMI), olanzapine (OLA), quetiapine (QUE), and aripiprazole (ARI) and enkephalinergic (ENK), substance P (SP), and tyrosine hydroxylase (TH) innervation fields in the rat septum. Olanzapine 213-216 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 31517634-1 2019 OBJECTIVE: The aim of the present study was to demonstrate the spatial relationship between the c-Fos immunoreactive cells elicited by an acute treatment with neuroleptics including amisulpride (AMI), olanzapine (OLA), quetiapine (QUE), and aripiprazole (ARI) and enkephalinergic (ENK), substance P (SP), and tyrosine hydroxylase (TH) innervation fields in the rat septum. Quetiapine Fumarate 219-229 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 31517634-1 2019 OBJECTIVE: The aim of the present study was to demonstrate the spatial relationship between the c-Fos immunoreactive cells elicited by an acute treatment with neuroleptics including amisulpride (AMI), olanzapine (OLA), quetiapine (QUE), and aripiprazole (ARI) and enkephalinergic (ENK), substance P (SP), and tyrosine hydroxylase (TH) innervation fields in the rat septum. Quetiapine Fumarate 231-234 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 31517634-1 2019 OBJECTIVE: The aim of the present study was to demonstrate the spatial relationship between the c-Fos immunoreactive cells elicited by an acute treatment with neuroleptics including amisulpride (AMI), olanzapine (OLA), quetiapine (QUE), and aripiprazole (ARI) and enkephalinergic (ENK), substance P (SP), and tyrosine hydroxylase (TH) innervation fields in the rat septum. Aripiprazole 241-253 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 31517634-1 2019 OBJECTIVE: The aim of the present study was to demonstrate the spatial relationship between the c-Fos immunoreactive cells elicited by an acute treatment with neuroleptics including amisulpride (AMI), olanzapine (OLA), quetiapine (QUE), and aripiprazole (ARI) and enkephalinergic (ENK), substance P (SP), and tyrosine hydroxylase (TH) innervation fields in the rat septum. Aripiprazole 255-258 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 30919007-5 2019 METHODS: First, we determined the effects of MA alone versus MA in combination with the dopamine receptor D1 antagonist SCH-23390 or the dopamine receptor D2 antagonist sulpiride on cFos expression and monoamines at the age when rats in the cognitive experiment were to begin testing and monoamines in rats after cognitive testing. Sulpiride 169-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 182-186 30919007-6 2019 RESULTS: SCH-23390 infused into the neostriatum prior to systemic administration of MA attenuated MA-induced cFos activation while sulpiride induced cFos activation. SCH 23390 9-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-113 30919007-6 2019 RESULTS: SCH-23390 infused into the neostriatum prior to systemic administration of MA attenuated MA-induced cFos activation while sulpiride induced cFos activation. Sulpiride 131-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 149-153 30991129-15 2019 In addition, because of the increase in the expression of fas and c-fos protein in testicle tissues, perchlorate could induce apoptosis in spermatogenesis. perchlorate 101-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 31248019-7 2019 The results of real-time PCR confirmed that DHA inhibits cerulein-induced IP3R1 and RyR2 gene expression, and demonstrated that DHA pre-treatment decreases the expression of the Relb gene, which encodes a component of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kappaB) transcriptional activator complex, and the c-fos gene, which encodes a component of activator protein-1 (AP-1) transcriptional activator complex. Docosahexaenoic Acids 128-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 340-345 31248019-8 2019 Taken together, DHA inhibits mRNA expression of IP3R1, RyR2, Relb, and c-fos, which is related to Ca2+ network in cerulein-stimulated PACs. Docosahexaenoic Acids 16-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 31281769-12 2019 After DMV destruction, c-fos expression was reduced in the AM, PVN, PBN, NTS/DMV, NA, RNM, and AP, especially in the AM, PVN, NTS/DMV, RNM, and AP. (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 6-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 31281769-12 2019 After DMV destruction, c-fos expression was reduced in the AM, PVN, PBN, NTS/DMV, NA, RNM, and AP, especially in the AM, PVN, NTS/DMV, RNM, and AP. (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 77-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 31281769-12 2019 After DMV destruction, c-fos expression was reduced in the AM, PVN, PBN, NTS/DMV, NA, RNM, and AP, especially in the AM, PVN, NTS/DMV, RNM, and AP. (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 77-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 30990106-4 2019 MATERIALS AND METHODS: MK801 (NMDA antagonist)-induced Fos-like immunoreactive (-LIR) neurons in contra-MVe, which had been suppressed by NMDA-mediated cerebellar inhibition in UL rats was used as an index. Dizocilpine Maleate 23-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 30990106-4 2019 MATERIALS AND METHODS: MK801 (NMDA antagonist)-induced Fos-like immunoreactive (-LIR) neurons in contra-MVe, which had been suppressed by NMDA-mediated cerebellar inhibition in UL rats was used as an index. N-Methylaspartate 30-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 30990106-4 2019 MATERIALS AND METHODS: MK801 (NMDA antagonist)-induced Fos-like immunoreactive (-LIR) neurons in contra-MVe, which had been suppressed by NMDA-mediated cerebellar inhibition in UL rats was used as an index. N-Methylaspartate 138-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 30990106-5 2019 RESULTS: The number of MK801-induced Fos-LIR neurons in contra-MVe gradually decreased to the same level as that of sham-operated rats 14 days after UL. Dizocilpine Maleate 23-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 30854919-12 2019 The fetal SCN is sensitive to GCs in vivo because DEX administration to pregnant rats acutely downregulates c-fos expression specifically in the laser-dissected fetal SCN. Dexamethasone 50-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 30935682-10 2019 Isoflurane anesthesia inhibited the activity of serotonergic neurons, as shown by decrease in the number of c-Fos-immunoreactive serotonergic neurons when compared with the sham group. Isoflurane 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 30861576-14 2019 Staining for c-fos was enhanced using the ABC method in that c-fos stained neurons were darker and more clearly visible after shorter treatment with DAB. 3,3'-Diaminobenzidine 149-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 30861576-14 2019 Staining for c-fos was enhanced using the ABC method in that c-fos stained neurons were darker and more clearly visible after shorter treatment with DAB. 3,3'-Diaminobenzidine 149-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 30905743-0 2019 c-Fos expression response to olanzapine, amisulpride, aripiprazole, and quetiapine single administration in the rat forebrain: Effect of a mild stress preconditioning. Olanzapine 29-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 30905743-0 2019 c-Fos expression response to olanzapine, amisulpride, aripiprazole, and quetiapine single administration in the rat forebrain: Effect of a mild stress preconditioning. Amisulpride 41-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 30905743-0 2019 c-Fos expression response to olanzapine, amisulpride, aripiprazole, and quetiapine single administration in the rat forebrain: Effect of a mild stress preconditioning. Aripiprazole 54-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 30905743-0 2019 c-Fos expression response to olanzapine, amisulpride, aripiprazole, and quetiapine single administration in the rat forebrain: Effect of a mild stress preconditioning. Quetiapine Fumarate 72-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 30998842-5 2019 Rats with lesser sucrose intake suppression in response to CCK gained more weight after HFHSD maintenance and displayed reduced CCK-induced c-Fos activation in the nucleus tractus solitarius. Sucrose 17-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 30990106-6 2019 Thioperamide moved the disappearance of the MK801-induced Fos-LIR neurons 2 days earlier. thioperamide 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 30990106-6 2019 Thioperamide moved the disappearance of the MK801-induced Fos-LIR neurons 2 days earlier. Dizocilpine Maleate 44-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 30990106-7 2019 The number of MK801-induced Fos-LIR neurons in thioperamide-treated rats was significantly decreased, compared with that of vehicle rats on days 7 and 12 after UL. Dizocilpine Maleate 14-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 30990106-7 2019 The number of MK801-induced Fos-LIR neurons in thioperamide-treated rats was significantly decreased, compared with that of vehicle rats on days 7 and 12 after UL. thioperamide 47-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 30990106-9 2019 CONCLUSION: These findings suggested that the number of MK801-induced Fos-LIR neurons in contra-MVe was decreased in concordance with the restoration of ipsi-MVe-activity during the late process of VC after UL and that thioperamide accelerated the late, but not the initial process of VC. Dizocilpine Maleate 56-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 30990106-9 2019 CONCLUSION: These findings suggested that the number of MK801-induced Fos-LIR neurons in contra-MVe was decreased in concordance with the restoration of ipsi-MVe-activity during the late process of VC after UL and that thioperamide accelerated the late, but not the initial process of VC. thioperamide 219-231 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 31159189-6 2019 Hypothalamic ghrelin sensitivity and microglia activation was evaluated using immunodetection for Iba-1 and c-Fos markers, and Western blot for TBK1 signaling. Ghrelin 13-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 31159189-8 2019 We found that programmed offspring by CAF diet exhibits overfeeding after fasting and peripheral ghrelin administration, which correlates with an increase in the hypothalamic Iba-1 microglia marker and c-Fos cell activation. cafestol palmitate 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 202-207 31159189-8 2019 We found that programmed offspring by CAF diet exhibits overfeeding after fasting and peripheral ghrelin administration, which correlates with an increase in the hypothalamic Iba-1 microglia marker and c-Fos cell activation. Ghrelin 97-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 202-207 31159189-13 2019 palmitic acid substantially activates hypothalamic Iba-1 microglia marker and c-Fos. Palmitic Acid 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 30703397-6 2019 At the molecular level, we found that metformin pretreatment not only prevented the changes of FOS, JUNB and BDNF at both mRNA and protein levels, but also increased the expression of the postsynaptic scaffold genes HOMER and PSD95 after exposure to hypobaric hypoxia. Metformin 38-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 30772369-7 2019 Striatal 6-OHDA induced motor impairments and reduced dopaminergic neuron immunolabeling as well as the pattern of neuronal activation (c-Fos) in the substantia nigra ipsilateral (IPL) to the lesion. Oxidopamine 9-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 31068813-8 2019 In addition, electroencephalograph (EEG) and electromyography (EMG) recordings of rats showed that loganin (35 mg/kg) prolonged the ratio of non-rapid eye movement (NREM) sleep and shortened wakefulness significantly, further immunohistochemistry showed that loganin (35 mg/kg) increased c-Fos expression in GABAergic neurons of rats in the ventrolateral preoptic nucleus (VLPO). loganin 99-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 288-293 30955316-10 2019 Compared with the saline group, intrathecal SB-366791 significantly decreased EMG activity (P<0.05) as well as spinal c-FOS (P<0.05) expression induced by UCD. Antimony 44-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-126 30169377-6 2019 The observed pattern of c-Fos expression suggests that the first exposure to the maze activates serotonin cells in the rostral and dorsal regions of the DRN and that only the dorsal subregion is activated by a second exposure. Serotonin 96-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 30388381-6 2019 Linoleic acid produced a greater increase in the mRNA expression of c-fos, c-jun, NF-kappaB, NFAT3, ANP, and BNP relative to palmitic acid and oleic acid. Linoleic Acid 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 30388381-6 2019 Linoleic acid produced a greater increase in the mRNA expression of c-fos, c-jun, NF-kappaB, NFAT3, ANP, and BNP relative to palmitic acid and oleic acid. Oleic Acid 3-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 30570802-9 2019 The number of spinal dorsal neurons co-expressed with EPAC1 and c-Fos after the incision was significantly lower in the propofol group. Propofol 120-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 29907842-7 2019 Sweet condensed milk (SCM) served as a reward to test consumption and subsequent activation (Fos+) of Nac and LHA neurons. nac 102-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 31013343-6 2019 Eye wipe behaviors in response to hypertonic saline and capsaicin were examined, and corneal mustard oil-induced c-Fos immunohistochemistry was quantified in the brainstem spinal trigeminal nucleus. mustard oil 93-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 30433807-9 2019 HCl perfusion and citric acid-induced cough caused Fos expressions in the nucleus of solitary tract (nTS), dorsal motor nucleus of the vagus (DMV), paratrigeminal nucleus (Pa5), and intermediate reticular nucleus (IRt), which was higher than HCl group, saline control group, citric acid-induced cough group, and blank group. Hydrochloric Acid 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 30433807-9 2019 HCl perfusion and citric acid-induced cough caused Fos expressions in the nucleus of solitary tract (nTS), dorsal motor nucleus of the vagus (DMV), paratrigeminal nucleus (Pa5), and intermediate reticular nucleus (IRt), which was higher than HCl group, saline control group, citric acid-induced cough group, and blank group. Citric Acid 18-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 30498893-8 2019 Here, we show that cocaine SA decreased PrL-NA core spine head diameter, nuclear Fos-IR and pCREB-IR, and GluA1-IR and GluA2-IR in putative mushroom-type spines 2 h after the end of cocaine SA, whereas the opposite occurred following 1 week of abstinence. cocaine sa 19-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-84 30790470-0 2019 Divergent effects of repeated cocaine and novel environment exposure on locus coeruleus c-fos expression and brain catecholamine concentrations in rats. Cocaine 30-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 28971699-11 2019 MK-8825 blocked c-fos induction by CSD in the brainstem trigeminal nucleus caudalis (TNC) and reticular nucleus of thalamus (TRN) but not in the amygdala. MK-8825 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 30571983-0 2019 Non-sulfated cholecystokinin-8 increases enteric and hindbrain Fos-like immunoreactivity in male Sprague Dawley rats. cholecystokinin 8 13-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 30571983-3 2019 We found that intraperitoneal NS CCK-8 (0.5 nmol/kg) increases Fos-LI in the DVC, the myenteric and the submucosal plexuses of the duodenum and the myenteric plexus of the jejunum. Sincalide 33-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 30604026-0 2019 Effect of orange juice and tryptamine on the behavior and c-fos expression of Wistar rats. tryptamine 27-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 30604026-4 2019 The aim of this study was to determine whether chronic administration of orange juice extract and tryptamine affects the behavior and c-fos expression in the rat. tryptamine 98-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-139 30729450-8 2019 In addition, paraoxon-induced Fos expression in the IO was also antagonized by MEC, but not by THP, and lesioning of the IO markedly suppressed tremorgenic action of paraoxon. Paraoxon 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 30934771-8 2019 Suppression of the heterodimeric combination of c-Jun and c-Fos proteins at the AP-1 binding site is highly involved in quercetin-mediated cytoprotection. Quercetin 120-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 31013343-10 2019 The number of Fos-positive neurons located in the caudal region of the spinal trigeminal nucleus after corneal mustard oil application was reduced in progesterone-treated animals. mustard oil 111-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 29877027-6 2019 Moreover, c-Fos quantification revealed that rats in the CD group showed hyperactivity in the lateral orbitofrontal cortex and basolateral amygdala when compared with the LD group. Cadmium 57-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-15 30594562-13 2019 Third, AS + ES of SSD rats resulted in bilateral reduced Fos expression in the auditory brainstem compared to monaural stimulations. as + es 7-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 30359643-4 2019 The goal of this study was to evaluate the acute action of ketamine on behavioural and neurochemical aspects of episodic-like memory (WWWhen/ELM task) through immediate-early gene expression (IEG), c-Fos, in the dorsal hippocampus. Ketamine 59-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 198-203 30359643-9 2019 In addition, the highest dose of ketamine increased c-Fos expression in dorsal CA1 subregion when compared to saline rats. Ketamine 33-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 30553789-10 2019 Moreover, intracerebroventricular injection with the PKC blocker chelerythrine chloride alleviated IS infusion-induced hyperalgesia and reduced ASIC3, CGRP and c-Fos levels in the TNC. chelerythrine 65-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 30723690-0 2019 Acute glufosinate-based herbicide treatment in rats leads to increased ocular interleukin-1beta and c-Fos protein levels, as well as intraocular pressure. phosphinothricin 6-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 30937353-8 2019 Compared to rats exposed to EPS only, significant reductions in acute stress-induced c-Fos expression were observed in the medial prefrontal cortex, hippocampus, and paraventricular nucleus of the hypothalamus (PVN) in CRS-EPS male rats, some of which persisted to 7 days post-stress. eps 28-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 30700692-7 2019 Our results also indicated that there was significantly lower expression of D1R, p-ERK1/2, and c-Fos in the IC of the U0126+Ket group, SCH23390+Ket group, and Ket+EA1 group as compared with that of the Ket group. U 0126 118-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 30700692-7 2019 Our results also indicated that there was significantly lower expression of D1R, p-ERK1/2, and c-Fos in the IC of the U0126+Ket group, SCH23390+Ket group, and Ket+EA1 group as compared with that of the Ket group. Ketamine 124-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 30700692-8 2019 CONCLUSIONS Ketamine addiction induces c-Fos overexpression in the IC by increasing the expression of D1R and p-ERK1/2. Ketamine 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 30587409-11 2019 Intradental capsaicin induced a significant increase in c-fos expression in the vlPAG that was exaggerated by orexin-A (0.51 mug/rat). Capsaicin 12-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 30587409-12 2019 Blockage of OX1R and CB1 receptors attenuated the effect of orexin-A on c-fos expression in capsaicin-treated rats. Capsaicin 92-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 31956557-3 2020 The aim of this study, was to analyze the behavioral effects of acute administration of a Mt infusion on male rats, during the Open Field (OFT) and Forced Swim (FST) Tests, and their association with the activation of oxytocin (OXT) cells, indicated by Fos protein (Fos/OXT) in the paraventricular (PVN) and supraoptic nuclei (SON). Oxytocin 218-226 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 253-256 31956557-3 2020 The aim of this study, was to analyze the behavioral effects of acute administration of a Mt infusion on male rats, during the Open Field (OFT) and Forced Swim (FST) Tests, and their association with the activation of oxytocin (OXT) cells, indicated by Fos protein (Fos/OXT) in the paraventricular (PVN) and supraoptic nuclei (SON). Oxytocin 218-226 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 266-269 30723690-7 2019 The results established that the glufosinate-based herbicide significantly increased IL-1beta and c-Fos immunopositivity in the optic nerve (p < 0.05), concomitant with increased IOP. phosphinothricin 33-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 30723838-9 2019 The 40 mg/kg ketamine infusion increased c-Fos levels in the mPFC and amygdala as well as pERK levels in the mPFC and hippocampus. Ketamine 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 31587016-0 2019 Differential expression of the c-fos protein and synaptophysin in zebrin II positive and zebrin II negative cerebellar cortical areas in 4-aminopyridine seizures. 4-Aminopyridine 137-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 30584976-10 2019 RESULTS: BDNF level increased while c-fos, Bax and caspase-3 levels decreased (p < 0.0001, in all) in the hippocampal neurons of the groups that were pre-treated with vitamin D before the administration of PTZ and KA, in comparison with the PTZ and KA groups. Vitamin D 170-179 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 30307667-8 2019 Fos expression results indicated that only the prelimbic cortex (PL) was activated by both heroin and cocaine cues; in contrast, no significant cue-induced neuronal activation was observed in other brain areas. Heroin 91-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 30307667-8 2019 Fos expression results indicated that only the prelimbic cortex (PL) was activated by both heroin and cocaine cues; in contrast, no significant cue-induced neuronal activation was observed in other brain areas. Cocaine 102-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 30359566-0 2019 Impaired chemoreflex correlates with decreased c-Fos in respiratory brainstem centers of the streptozotocin-induced Alzheimer"s disease rat model. Streptozocin 93-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 30662294-10 2019 In addition, increase in c-Fos-positive cells within L4 dorsal horn lamina I and II of the STZ control group was markedly alleviated by GS-KG9. Streptozocin 91-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 30484727-0 2019 Adolescent cocaine exposure enhanced negative affect following drug re-exposure in adult rats: Attenuation of c-Fos activation. Cocaine 11-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 30484727-4 2019 In this regard, the results showed that adolescent cocaine exposure did not modulate cell proliferation (Ki-67+ cells) or c-Fos protein activation in the dentate gyrus region of the hippocampus, but attenuated c-Fos activation in the dorsal striatum. Cocaine 51-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 210-215 30458222-0 2019 Brain Sub/Region-Specific Effects of Olanzapine on c-Fos Expression of Chronically Socially Isolated Rats. Olanzapine 37-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 30419270-0 2019 The offspring of rats selected for high or low ethanol intake at adolescence exhibit differential ethanol-induced Fos immunoreactivity in the central amygdala and in nucleus accumbens core. Ethanol 47-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 30419270-0 2019 The offspring of rats selected for high or low ethanol intake at adolescence exhibit differential ethanol-induced Fos immunoreactivity in the central amygdala and in nucleus accumbens core. Ethanol 98-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 30419270-7 2019 STDRLO, but not STDRHI, rats exhibited ethanol-induced Fos-ir in Ce. Ethanol 39-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 30419270-8 2019 STRDHI, but not STDRLO, rats exhibited an ethanol-induced Fos-ir depression in AcbC. Ethanol 42-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 30419270-8 2019 STRDHI, but not STDRLO, rats exhibited an ethanol-induced Fos-ir depression in AcbC. 1-aminocyclobutanecarboxylic acid 79-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 30419270-11 2019 These results indicate that short term selection for differential alcohol intake during adolescence yields heightened neural activity at baseline (i.e., after vehicle) in STRDHI vs. STDRLO adolescent rats, and differential sensitivity to ethanol-induced Fos immunoreactivity in Ce and in AcbC. Alcohols 66-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 254-257 30419270-11 2019 These results indicate that short term selection for differential alcohol intake during adolescence yields heightened neural activity at baseline (i.e., after vehicle) in STRDHI vs. STDRLO adolescent rats, and differential sensitivity to ethanol-induced Fos immunoreactivity in Ce and in AcbC. Ethanol 238-245 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 254-257 30943499-11 2019 The protein levels of c-Fos and NGF were decreased by treatment with curcumin compared with the corresponding protein levels in the BPA group. Curcumin 69-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 30943499-11 2019 The protein levels of c-Fos and NGF were decreased by treatment with curcumin compared with the corresponding protein levels in the BPA group. bisphenol A 132-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 30973107-0 2019 The Effect of Alpha-Tocopherol on Morphine Tolerance-induced Expression of c-fos Proto-oncogene from a Biotechnological Perspective. alpha-Tocopherol 14-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 30973107-0 2019 The Effect of Alpha-Tocopherol on Morphine Tolerance-induced Expression of c-fos Proto-oncogene from a Biotechnological Perspective. Morphine 34-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 30973107-13 2019 Furthermore, we found that the Alpha-Tocopherol obviously decreased c-fos gene expression, especially in the spinal cord. alpha-Tocopherol 31-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 30856132-7 2019 The number of c-Fos-positive cells was elevated in the L4 and L5 segments; and on the contralesional compared to the ipsilesional side in the CCI group. CCI 142-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 30355627-6 2018 Both shock-induced reinstatement and the prelimbic cortex Fos response were prevented by bilateral intra-VTA injections of the CRF receptor 1 (CRFR1) antagonist, antalarmin. antalarmin 162-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 30359566-11 2019 STZ-AD rats showed a significant decrease of c-Fos labeling in the caudal/medial nTS, rostral ventral respiratory group, and Botzinger complex. Streptozocin 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 30359566-14 2019 This decrease in c-Fos expression correlates with the observed blunting of respiration to hypoxia in the STZ-AD rat model. Streptozocin 105-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 30286535-5 2018 The results showed that after treatment with DnBP (20 mumol/L), cell proliferation increased to 114.69% (p < 0.01) and c-fos gene was up-regulated by 1.57-fold (p < 0.01). Dibutyl Phthalate 45-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 30286535-8 2018 Finally, DnBP activated the downstream extracellular regulated protein kinases (ERK1/2) pathway, up-regulating Mapk1 (1.15-, p < 0.05), Mapk3 (1.26-fold, p < 0.01) and increasing protein levels of p-ERK (p < 0.01); notably, DnBP-induced ERK1/2 activation along with c-fos up-regulation were attenuated by PD98059 (ERK1/2 inhibitor). Dibutyl Phthalate 9-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 275-280 30242293-4 2018 In the short-term, at 30 days of age, the respiratory frequency (RF) and immunoreactivity for Fos on the retrotrapezoid nucleus (RTN; brainstem site containing CO2 sensitive neurons) after exposure to CO2 were evaluated. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 201-204 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 30085976-8 2018 c-Fos mapping in feeding-related brain sites revealed TRP-induced changes in the dorsal vagal complex, hypothalamic paraventricular and supraoptic nuclei and in the basolateral amygdala. Tryptophan 54-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 30014576-8 2018 The c-Fos expression analysis indicated that neuronal activity and the number of neurons in the dorsal raphe nucleus and locus coeruleus decreased in STZ-injected rats. Streptozocin 150-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 29928872-3 2018 Chronic infusion of aldosterone (100 ng/h) into the 4th V increased daily 0.3 M NaCl intake (up to 44 +- 10, vs. vehicle: 5.6 +- 3.4 ml/24 h) and also c-Fos expression in HSD2 neurons in the NTS and in non-HSD2 neurons in the NTS. Aldosterone 20-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-156 30318755-4 2018 We have previously shown that methyl supplementation via L-Methionine (MET) administration attenuates cocaine-seeking behavior and reverses expression and methylation patterns of the immediate early gene c-fos, suggesting that MET may act by altering the excitability of this circuitry during cocaine reinstatement. Methionine 57-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 204-209 30318755-4 2018 We have previously shown that methyl supplementation via L-Methionine (MET) administration attenuates cocaine-seeking behavior and reverses expression and methylation patterns of the immediate early gene c-fos, suggesting that MET may act by altering the excitability of this circuitry during cocaine reinstatement. Methionine 71-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 204-209 30136719-11 2018 In the dorsal motor nucleus of the vagus (DMV), insulin evoked Fos in many ChAT-positive neurons. (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 42-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 30136719-14 2018 The high numbers of PNMT-immunoreactive and ChAT-immunoreactive neurons that express Fos after insulin treatment reinforce the importance of these neurons in the central response to a decrease in glucose bioavailability. Glucose 196-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 29472642-10 2018 TAAR1 activation was sufficient to block nicotine-induced c-Fos expression in the NAc, while also reducing nicotine-induced dopamine release in the NAc. Nicotine 41-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 30403605-7 2018 Bax and C-fos were positively expressed and significantly higher in the hippocampus of chronic cerebral ischemic vascular dementia rats (model group) than in the medicated thread moxibustion group after treatment (p &lt;0.01). Phosphorus 19-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-13 30403605-7 2018 Bax and C-fos were positively expressed and significantly higher in the hippocampus of chronic cerebral ischemic vascular dementia rats (model group) than in the medicated thread moxibustion group after treatment (p &lt;0.01). Adenosine Monophosphate 78-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-13 30337738-16 2018 CONCLUSION: Minocycline can inhibit formalin-induced inflammatory pain and the expression of c-Fos protein in spinal dorsal horn. Minocycline 12-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 30201258-8 2018 RESULTS: Consumption of Splenda provoked an inverted U-shaped dose-effect in c-Fos expression in ventromedial hypothalamic nucleus while similar findings were observed in dentate gyrus of hippocampus. trichlorosucrose 24-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 30144148-0 2018 Clozapine impact on c-Fos expression in mild stress preconditioned male rats exposed to a novelty stressor. Clozapine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 30144148-7 2018 After a single FSW, CLZ decreased the number of c-Fos immunoreactive cells in the vLS, DMStr, NAc shell, and NAc core. Clozapine 20-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 30144148-7 2018 After a single FSW, CLZ decreased the number of c-Fos immunoreactive cells in the vLS, DMStr, NAc shell, and NAc core. nac 94-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 30485908-9 2018 In the dopamine-depleted striatum, Vilazodone-l-dopa cotreatment increased dopamine content, suggesting a normalization of dopamine kinetics in dyskinetic brain, and reduced l-dopa-induced c-Fos and preprodynorphin mRNA overexpression, indicative of attenuated dopamine D1 receptor-mediated direct pathway overactivity. Dopamine 7-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 189-194 30485908-9 2018 In the dopamine-depleted striatum, Vilazodone-l-dopa cotreatment increased dopamine content, suggesting a normalization of dopamine kinetics in dyskinetic brain, and reduced l-dopa-induced c-Fos and preprodynorphin mRNA overexpression, indicative of attenuated dopamine D1 receptor-mediated direct pathway overactivity. vilazodone-l-dopa 35-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 189-194 30485908-9 2018 In the dopamine-depleted striatum, Vilazodone-l-dopa cotreatment increased dopamine content, suggesting a normalization of dopamine kinetics in dyskinetic brain, and reduced l-dopa-induced c-Fos and preprodynorphin mRNA overexpression, indicative of attenuated dopamine D1 receptor-mediated direct pathway overactivity. Levodopa 46-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 189-194 30375397-4 2018 Suppression of fear in the extinction context is associated with an increase in c-fos expression and spike firing in RE neurons to the extinguished CS. Cesium 148-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 30170041-7 2018 The numbers of c-Fos-immunopositive cells with or without calbindin-D28K-immunoreactivity increased significantly in the MPNc and CALB-SDN following ejaculation in both sexually inexperienced and experienced males, although the numbers did not change significantly with exposure to estrous female odors, the first mount, and the first intromission before and after the first ejaculation. mpnc 121-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 30170041-7 2018 The numbers of c-Fos-immunopositive cells with or without calbindin-D28K-immunoreactivity increased significantly in the MPNc and CALB-SDN following ejaculation in both sexually inexperienced and experienced males, although the numbers did not change significantly with exposure to estrous female odors, the first mount, and the first intromission before and after the first ejaculation. calb-sdn 130-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 30058183-6 2018 Neural activation (via Fos protein) was quantified in the nucleus accumbens core (NAC) and shell (NAS). nac 82-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 30119205-4 2018 Our in vitro studies showed that TB significantly suppressed RANKL-induced osteoclastogenesis and the expression of related marker proteins, including NFATc1, TRAP, c-Fos, and cathepsin K. theabrownin 33-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 30170082-5 2018 We found that two-week FLX treatment dramatically inhibited Fos expression in serotonin neurons and that this effect was reversed by blocking 5-HT1A receptors with WAY. Fluoxetine 23-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 30310305-7 2018 The time spent on rubbing directed to the injected area and c-Fos immunoreactivity in TNC was measured as the formalin-induced pain-related behavior, and as the marker of pain-related neuronal activation, respectively. Formaldehyde 110-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 30069980-9 2018 Water avoidance stress-derived mucosal supernatants markedly enhanced the c-fos expression and enteric synaptic plasticity in LMMP tissues, which could be eliminated by mast cell inhibition or NGF neutralization, but not tryptase or histamine blocking. Water 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 30139246-10 2018 The analgesic, gabapentin, reversed the mechanical hyper-sensitivity and the augmentedexpression of limbic pERK and c-Fos in the SNI rats. Gabapentin 15-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 30319425-7 2018 Results: In the VPA-treated group, the central responsiveness expressed by reducing the number of c-Fos-ir neurons, which had been increased by i.c. Valproic Acid 16-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 30241394-0 2018 Sodium Citrate Increases Expression and Flux of Mg2+ Transport Carriers Mediated by Activation of MEK/ERK/c-Fos Pathway in Renal Tubular Epithelial Cells. Sodium Citrate 0-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 30241394-0 2018 Sodium Citrate Increases Expression and Flux of Mg2+ Transport Carriers Mediated by Activation of MEK/ERK/c-Fos Pathway in Renal Tubular Epithelial Cells. magnesium ion 48-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 30241394-6 2018 SC increased the levels of p-ERK1/2 and p-c-Fos, which were inhibited by U0126. U 0126 73-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 29874094-0 2018 Activation of catecholamine neurons in the ventral medulla reduces CCK-induced hypophagia and c-Fos activation in dorsal medullary catecholamine neurons. Catecholamines 14-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 29874094-0 2018 Activation of catecholamine neurons in the ventral medulla reduces CCK-induced hypophagia and c-Fos activation in dorsal medullary catecholamine neurons. Catecholamines 131-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 29874094-10 2018 Selective activation of A1/C1 neurons using the DREADD agonist, clozapine- N-oxide, attenuated reduction of food intake by CCK and prevented CCK-induced c-Fos expression in A2 CA neurons, even under normoglycemic conditions. clozapine N-oxide 64-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-158 30170186-8 2018 In conclusion, the results presented here provide compelling evidence of the modulation and involvement of the c-fos and the CREB molecules in the cannabinoid-opioid interaction of the brain reward system in the CPP paradigm. Cannabinoids 147-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 29194604-5 2018 The targeting relationship between miR-129 and c-Fos was predicted and verified by bioinformatics websites and dual-luciferase reporter gene assay. mir-129 35-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 29194604-9 2018 Compared with the blank and NC groups, the miR-129 mimic group and the siRNA-c-Fos group showed decreased expression of c-Fos, Bax, and MAPK, increased cells proliferation, and decreased cell apoptosis, fewer cells arrested in the G1 phase and more cells arrested in the S phase. mir-129 43-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 29194604-11 2018 This study suggests that miR-129 could inhibit the occurrence and development of epilepsy by repressing c-Fos expression through inhibiting the MAPK signaling pathway. mir-129 25-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 30140065-9 2018 Greater numbers of c-Fos-positive cells in the rostral ventrolateral medulla (RVLM) and of c-Fos-positive orexin neurons in the dorsomedial hypothalamus (DMH) were detected in sucrose agar-treated SHRs, compared to regular chow-treated SHRs and to sucrose agar-treated WKY rats. 1,2-Dimethylhydrazine 154-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 30140065-9 2018 Greater numbers of c-Fos-positive cells in the rostral ventrolateral medulla (RVLM) and of c-Fos-positive orexin neurons in the dorsomedial hypothalamus (DMH) were detected in sucrose agar-treated SHRs, compared to regular chow-treated SHRs and to sucrose agar-treated WKY rats. Sucrose 176-183 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 30140065-9 2018 Greater numbers of c-Fos-positive cells in the rostral ventrolateral medulla (RVLM) and of c-Fos-positive orexin neurons in the dorsomedial hypothalamus (DMH) were detected in sucrose agar-treated SHRs, compared to regular chow-treated SHRs and to sucrose agar-treated WKY rats. Sucrose 176-183 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 29669266-4 2018 When animals received scopolamine or MK-801 treatment prior to conditioning sessions we observed significantly fewer TH-labeled (i.e., DA) cells in the VTA that expressed c-fos and significantly less conditioned approach responding during the CS-only test. Scopolamine 22-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-176 29669266-4 2018 When animals received scopolamine or MK-801 treatment prior to conditioning sessions we observed significantly fewer TH-labeled (i.e., DA) cells in the VTA that expressed c-fos and significantly less conditioned approach responding during the CS-only test. Dizocilpine Maleate 37-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-176 29952605-5 2018 An elevation of Fos-like immunoreactivity in Ox neurons was observed in fluid-depleted rats that were allowed to ingest water and sodium. Water 120-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 29952605-5 2018 An elevation of Fos-like immunoreactivity in Ox neurons was observed in fluid-depleted rats that were allowed to ingest water and sodium. Sodium 130-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 29808228-2 2018 Two experiments were conducted to assess: (1) whether repeated forced swim stress for 3 days increased the number of Fos-positive neurons evoked by masseter muscle injury due to formalin injection; and (2) whether serotonin-reuptake inhibitor, fluoxetine, administered daily after each stress conditioning, had modulatory roles on Fos expression. Formaldehyde 178-186 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 29808228-5 2018 Repeated forced swim stress significantly increased Fos expression in all Vc areas compared with those of non-stressed rats, while systemic administration of fluoxetine significantly decreased Fos expression in all areas, but mainly in the caudal Vc region, in stressed rats. Fluoxetine 158-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-196 29623778-5 2018 Moreover, double immunofluorescence was employed to evaluate the qualitative and quantitative expression of dopamine/ c-Fos in CRD rats. Dopamine 108-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 29934352-9 2018 Using Fluoro-Gold tracing and immunohistochemistry for c-Fos and Foxp2, a marker of sodium-deprivation responsive neurons, we revealed brainstem populations of neurons that are activated by sodium depletion and project directly to the ventral tegmental area. Sodium 190-196 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 29926762-0 2018 The galanin receptor-3 antagonist, SNAP 37889, inhibits cue-induced reinstatement of alcohol-seeking and increases c-Fos expression in the nucleus accumbens shell of alcohol-preferring rats. Alcohols 166-173 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 29926762-1 2018 INTRODUCTION: This study aimed to investigate the effects of the galanin-3 receptor antagonist, SNAP 37889, on c-Fos protein expression after cue-induced reinstatement of alcohol-seeking in the brains of alcohol-preferring rats. Alcohols 171-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 29926762-1 2018 INTRODUCTION: This study aimed to investigate the effects of the galanin-3 receptor antagonist, SNAP 37889, on c-Fos protein expression after cue-induced reinstatement of alcohol-seeking in the brains of alcohol-preferring rats. Alcohols 204-211 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 29926762-7 2018 RESULTS: SNAP 37889 administration increased c-Fos immunoreactivity in the nucleus accumbens shell, but was without effect in the nucleus accumbens core and the medial prefrontal cortex. 1-phenyl-3-((3-(trifluoromethyl)phenyl)imino)-1H-indol-2-one 9-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 30003352-10 2018 RESULTS: NTG administration induced spinal hyperalgesia and orofacial allodynia, together with a significant increase in the expression of CGRP and c-Fos genes in trigeminal ganglia and central areas. Nitroglycerin 9-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 29518407-5 2018 Fos immunolabeling in response to isoproterenol was reduced in both the SFO and the OVLT of aged females compared to young females, but was increased in the SON of female rats of both ages. Isoproterenol 34-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 30003352-11 2018 Topiramate treatment prevented NTG-induced changes by reversing NTG-induced hyperalgesia and allodynia, and inhibiting CGRP and c-Fos gene expression in all areas evaluated. Topiramate 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-133 29977067-1 2018 Previous studies showed that restraint water-immersion stress (RWIS) increases the expression of Fos protein in the ventromedial hypothalamic nucleus (VMH), indicating the VMH involving in the stress-induced gastric mucosal injury (SGMI). Water 39-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 28616820-3 2018 Fos-like immunoreactivity (Fos-LI) expressed in the supraoptic nucleus (SON), the paraventricular nucleus (PVN), the area postrema and the nucleus of the solitary tract (NTS) after intraperitoneal (ip) administration of cisplatin. Cisplatin 220-229 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 28616820-3 2018 Fos-like immunoreactivity (Fos-LI) expressed in the supraoptic nucleus (SON), the paraventricular nucleus (PVN), the area postrema and the nucleus of the solitary tract (NTS) after intraperitoneal (ip) administration of cisplatin. Cisplatin 220-229 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 28616820-7 2018 In the OXT-LI cells, cisplatin-induced Fos expression in the SON and the PVN was also suppressed by OXTR-A pretreatment. Cisplatin 21-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 29441405-7 2018 In rats that reinstated methamphetamine seeking, these altered electrophysiological properties of GCNs were associated with enhanced expression of Fos, GluN2A subunits and PSD95 and reduced expression of GABAA subunits in the DG and enhanced expression of synaptic PSD in the molecular layer. Methamphetamine 24-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-150 29671014-7 2018 Fos in OXT neurons was further decreased on ED1, compared to homecage controls, in both males and females even though in SON, cocaine exposure increased the number of OXT-expressing neurons. Cocaine 126-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 29705538-5 2018 The increased Fos expression in the T11 and T12, but not T8 - T10 spinal cord segments, was significantly blocked by local application of either N-methyl-D-aspartate (NMDA) receptor antagonist dizocilpine maleate (MK-801) or non-NMDA receptor antagonist 6,7-dinitroquinoxaline-2,3-dione (DNQX) into the receptive field of T11 CBDR. D-Aspartic Acid 154-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 29705538-5 2018 The increased Fos expression in the T11 and T12, but not T8 - T10 spinal cord segments, was significantly blocked by local application of either N-methyl-D-aspartate (NMDA) receptor antagonist dizocilpine maleate (MK-801) or non-NMDA receptor antagonist 6,7-dinitroquinoxaline-2,3-dione (DNQX) into the receptive field of T11 CBDR. Dizocilpine Maleate 193-212 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 29705538-5 2018 The increased Fos expression in the T11 and T12, but not T8 - T10 spinal cord segments, was significantly blocked by local application of either N-methyl-D-aspartate (NMDA) receptor antagonist dizocilpine maleate (MK-801) or non-NMDA receptor antagonist 6,7-dinitroquinoxaline-2,3-dione (DNQX) into the receptive field of T11 CBDR. Dizocilpine Maleate 214-220 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 29705538-5 2018 The increased Fos expression in the T11 and T12, but not T8 - T10 spinal cord segments, was significantly blocked by local application of either N-methyl-D-aspartate (NMDA) receptor antagonist dizocilpine maleate (MK-801) or non-NMDA receptor antagonist 6,7-dinitroquinoxaline-2,3-dione (DNQX) into the receptive field of T11 CBDR. FG 9041 254-286 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 29705538-5 2018 The increased Fos expression in the T11 and T12, but not T8 - T10 spinal cord segments, was significantly blocked by local application of either N-methyl-D-aspartate (NMDA) receptor antagonist dizocilpine maleate (MK-801) or non-NMDA receptor antagonist 6,7-dinitroquinoxaline-2,3-dione (DNQX) into the receptive field of T11 CBDR. FG 9041 288-292 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 29705538-5 2018 The increased Fos expression in the T11 and T12, but not T8 - T10 spinal cord segments, was significantly blocked by local application of either N-methyl-D-aspartate (NMDA) receptor antagonist dizocilpine maleate (MK-801) or non-NMDA receptor antagonist 6,7-dinitroquinoxaline-2,3-dione (DNQX) into the receptive field of T11 CBDR. cbdr 326-330 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 29928003-8 2018 Consequentially, after electrical stimulation of the CCI-treated median nerve, the number of c-Fos-LI neurons in the cuneate nucleus (CN) was significantly reduced in the M871 group, whereas it increased in the AR-M1896 group. CCI 53-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 29928003-9 2018 These results suggest that activation of GalR2, probably through NPY or nitric oxide, induces c-Fos expression in the CN and transmits mechanical allodynia sensations to the thalamus. Nitric Oxide 72-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 29923357-7 2018 DENA administration activated EGFR/ERK1/2 signaling and caused a significant increase in P-EGFR and P-ERK1/2 as well as a significant up-regulation of expression of target genes such as PCNA, c-fos and Bcl2, which indicated the increase in cell proliferation. Diethylnitrosamine 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 192-197 29344989-10 2018 Likewise, GTN-induced c-fos expression in TNC. Nitroglycerin 10-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 29524326-11 2018 Anaphylaxis increased c-fos expression in the neurons of the paraventricular nucleus (PVN) of the hypothalamus and in those of the nucleus tractus solitarii (NTS) and rostral ventrolateral medulla (RVLM) of the medulla oblongata; c-fos was expressed in gamma-aminobutyric acid (GABA)-ergic neurons of the NTS and in the catecholaminergic neurons of the RVLM. gamma-Aminobutyric Acid 253-276 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 29524326-11 2018 Anaphylaxis increased c-fos expression in the neurons of the paraventricular nucleus (PVN) of the hypothalamus and in those of the nucleus tractus solitarii (NTS) and rostral ventrolateral medulla (RVLM) of the medulla oblongata; c-fos was expressed in gamma-aminobutyric acid (GABA)-ergic neurons of the NTS and in the catecholaminergic neurons of the RVLM. gamma-Aminobutyric Acid 278-282 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 29528526-13 2018 Minocycline treatment also elicited a decrease in the number of activated neurons in the RVLM/C1 neurons (expressed as Fos+ /tyrosine hydroxylase+ ), the number of Fos-activated neurons in the PVH and the increase in ventilation elicited by AH. Minocycline 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-122 29528526-13 2018 Minocycline treatment also elicited a decrease in the number of activated neurons in the RVLM/C1 neurons (expressed as Fos+ /tyrosine hydroxylase+ ), the number of Fos-activated neurons in the PVH and the increase in ventilation elicited by AH. Minocycline 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-167 30013938-9 2018 DEN effectively activated apoptotic markers GSK-3 and c-FOS. Diethylnitrosamine 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 29450645-7 2018 Histone 3 phosphorylation at serine 10, another neuronal activation marker, corroborated c-fos results. Serine 29-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 29896090-8 2018 Strikingly, c-Fos immunohistochemistry revealed that while voluntary running activity elevated the number of c-Fos positive cells in the DMH (particularly in the ventral and caudal subregions) of intact rats, such activation was not observed in ZF rats. 1,2-Dimethylhydrazine 137-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 29896090-8 2018 Strikingly, c-Fos immunohistochemistry revealed that while voluntary running activity elevated the number of c-Fos positive cells in the DMH (particularly in the ventral and caudal subregions) of intact rats, such activation was not observed in ZF rats. 1,2-Dimethylhydrazine 137-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 29861656-5 2018 c-Fos immunofluorescence of the brain stem showed that dorsal motor nucleus of the vagus was activated after remifentanil preconditioning. Remifentanil 109-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 29496477-8 2018 Following CNN, acute nicotine stimulated IEG expression in all three areas, but activation was significantly reduced in the LC (c-Fos, Egr-1, Npas4), and CeA (c-Fos). Nicotine 21-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-133 29496477-8 2018 Following CNN, acute nicotine stimulated IEG expression in all three areas, but activation was significantly reduced in the LC (c-Fos, Egr-1, Npas4), and CeA (c-Fos). Nicotine 21-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-164 29567624-6 2018 Next, we determined the effect of context-induced renewal of alcohol-seeking behavior on the expression of Fos (a neuronal activity marker) in the OFC. Alcohols 61-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 28929435-11 2018 Neuroglial changes, c-Fos, and protein oxidation levels significantly reduced after galantamine and galantamine plus atropine treatment indicate their potential antidotal value in nerve agent treatment. Galantamine 84-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 29574216-7 2018 ABA increased the number of c-Fos/CRF double labeled neurons in the paraventricular nucleus (PVN) and the dorsomedial hypothalamic nucleus (DMH) compared to the ad libitum (AL, ad libitum fed, no running wheel) and activity (AC, ad libitum fed, running wheel, p < 0.05) but not to the restricted feeding (RF, food for 1.5 h/day, no running wheel, p > 0.05) group. alisol B 23-acetate 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 29574216-9 2018 In the Edinger-Westphal nucleus (EW) the number of c-Fos positive neurons was increased in ABA and RF compared to AC (p < 0.05), while the number of double labeled neurons was not different (p > 0.05). alisol B 23-acetate 91-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 29212295-4 2018 In alveolar bone, histological analysis with staining and micro-computed tomography revealed the attenuation of alveolar bone resorption in the BPE-treated aged group, which led to a significant reduction in the mRNA and protein expression of nuclear factor of activated T-cells c1 (NFATc1), c-Fos, tartrate-resistant acid phosphatase, and cathepsin K (p < 0.01). 1,2-bis(4-pyridyl)ethene 144-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 292-297 29292158-2 2018 Using the single prolonged stress (SPS) model of post traumatic stress disorder and the immediate early gene (IEG) c-Fos as a measure of neural activity, we previously identified patterns of neural activity through which SPS disrupts extinction retention. Sodium phenolsulfonate 221-224 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 29292158-4 2018 C-Jun is another IEG that is sometimes regulated in a different manner to c-Fos and could be used to identify emotional learning/memory specific patterns of neural activity that are sensitive to SPS. Sodium phenolsulfonate 195-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 28730575-4 2018 Results showed that as the fluoride concentration increased, calcium ion concentration [Ca2+], the expression of calcium/calmodulin-dependent protein kinase II alpha (CaMKIIalpha), and the expression of catus proto-oncogene protein c-fos (c-fos) all tend to increase. Fluorides 27-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 232-237 28730575-4 2018 Results showed that as the fluoride concentration increased, calcium ion concentration [Ca2+], the expression of calcium/calmodulin-dependent protein kinase II alpha (CaMKIIalpha), and the expression of catus proto-oncogene protein c-fos (c-fos) all tend to increase. Fluorides 27-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 239-244 28730575-5 2018 Compared to the control group, Ca2+, CaMKIIalpha, and c-fos significantly increased (P < 0.05) in the moderate-fluoride and the high-fluoride groups. Fluorides 114-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 28730575-5 2018 Compared to the control group, Ca2+, CaMKIIalpha, and c-fos significantly increased (P < 0.05) in the moderate-fluoride and the high-fluoride groups. Fluorides 136-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 28730575-6 2018 These results indicate that Ca2+/CaMKIIalpha/c-fos channel signal may be the molecular mechanism of central nervous system damage caused by chronic fluoride intoxication. Fluorides 148-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 29499191-0 2018 L-carnitine mitigates UVA-induced skin tissue injury in rats through downregulation of oxidative stress, p38/c-Fos signaling, and the proinflammatory cytokines. Carnitine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 29715183-0 2018 c-Fos expression in the hypothalamic paraventricular nucleus after a single treatment with a typical haloperidol and nine atypical antipsychotics: a pilot study. Haloperidol 101-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 29094451-10 2018 3) Cisplatin significantly increased the expression of c-fos in the NTS and AP. Cisplatin 3-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 29094451-12 2018 EA reduced the cisplatin-induced c-fos expression in the AP but not the NTS. Cisplatin 15-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 29355671-9 2018 Our data showed the ratio of pCREB/CREB, pP70/P70 and c-fos expression in the hippocampus significantly decreased after IPN reversible inactivation. ipn 120-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 29421187-4 2018 Brief exposure of rats to ether anesthesia evokes pathological burst firing and associated expression of the immediate early gene c-Fos in STN neurons. Ether 26-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 29421187-9 2018 Ether-evoked expression of c-Fos immunoreactivity was suppressed by HFS at 200 and 260 Hz with a pulse width of 60 micros, and by 130 Hz when the pulse width was increased to 90 micros. Ether 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 29650024-7 2018 Moreover, approximately 34.0% of the collateral projection neurons in the Vc showed Fos immunopositivity after injection of formalin into the lip, and parts of calcitonin gene-related peptide (CGRP)-immunopositive axonal varicosities were in direct contact with the Vc collateral projection neurons. Formaldehyde 124-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 29288824-8 2018 In the contextual-fear test, essential oil at 2.5% and 5% (but not 1%) reduced the freezing response and its respective c-Fos expression in the ventral hippocampus and amygdala. Oils, Volatile 29-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 29288824-10 2018 However, rats that inhaled essential oil at 2.5% and 5% (but not 1%) showed decreased freezing in the three minutes after tone presentation, as well as reduced c-Fos expression in the prefrontal cortex and amygdala. Oils, Volatile 27-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 28054308-4 2018 A marked increase was observed in the number of neurons showing Fos immunoreactivity in the paraventricular nucleus, arcuate nucleus and lateral hypothalamic area (LHA), 90 min after icv administration of ghrelin. Ghrelin 205-212 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 29371098-6 2018 Intra-oral infusion of dH2O, NaCl, or sucrose altered the number of Fos-IR neurons in only specific subareas of the GC and the effects of these tastants were concentration-dependent. dh2o 23-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 29371098-6 2018 Intra-oral infusion of dH2O, NaCl, or sucrose altered the number of Fos-IR neurons in only specific subareas of the GC and the effects of these tastants were concentration-dependent. Sodium Chloride 29-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 29371098-6 2018 Intra-oral infusion of dH2O, NaCl, or sucrose altered the number of Fos-IR neurons in only specific subareas of the GC and the effects of these tastants were concentration-dependent. Sucrose 38-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 29371098-6 2018 Intra-oral infusion of dH2O, NaCl, or sucrose altered the number of Fos-IR neurons in only specific subareas of the GC and the effects of these tastants were concentration-dependent. tastants 144-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 29371098-7 2018 For example, 1.0 M NaCl increased Fos-IR neurons in the posterior lateral AID and DI and elicited more aversive TR responses than 0.1 M NaCl. Sodium Chloride 19-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 29371098-8 2018 Compared to dH2O, infusions of HCl or QHCl increased the total number of Fos-IR neurons in many subareas of the GC throughout its anterior-posterior extent and increased aversive TR behaviors. Hydrochloric Acid 31-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-76 29371098-8 2018 Compared to dH2O, infusions of HCl or QHCl increased the total number of Fos-IR neurons in many subareas of the GC throughout its anterior-posterior extent and increased aversive TR behaviors. qhcl 38-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-76 28952321-0 2018 Cilostazol induces C-fos expression in the trigeminal nucleus caudalis and behavioural changes suggestive of headache with the migraine-like feature photophobia in female rats. Cilostazol 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 28952321-3 2018 We aimed to investigate whether cilostazol could provoke headache-like behaviours and c-fos expression in rats. Cilostazol 32-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 28952321-10 2018 Cilostazol also induced c-fos expression in the trigeminal nucleus caudalis. Cilostazol 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 29609419-10 2018 c-Fos, NGF, and NOS expression was suppressed by the combination treatment (tamsulosin with naftopidil) to a greater extent than was the case for tamsulosin monotherapy or naftopidil monotherapy. Tamsulosin 76-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 29609419-10 2018 c-Fos, NGF, and NOS expression was suppressed by the combination treatment (tamsulosin with naftopidil) to a greater extent than was the case for tamsulosin monotherapy or naftopidil monotherapy. naftopidil 92-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 29200178-8 2018 C-Fos expression in response to touch was increased in trigeminal nucleus caudalis in animals exposed to acrolein compared with room air. Acrolein 105-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 28589966-8 2018 U50,488-induced reinstatement of alcohol seeking was associated with increased Fos expression in multiple brain areas, including the BNST, where it was significantly correlated with lever pressing. Alcohols 33-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 29204804-4 2018 OBJECTIVES: This study examined whether manipulation of E2 in the mPOA modulates differing behavioral responses to cocaine and whether this is reflected in differing levels of c-Fos in the NAc following cocaine administration. nac 189-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 176-181 29204804-4 2018 OBJECTIVES: This study examined whether manipulation of E2 in the mPOA modulates differing behavioral responses to cocaine and whether this is reflected in differing levels of c-Fos in the NAc following cocaine administration. Cocaine 203-210 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 176-181 29204804-10 2018 Finally, animals that received cocaine had increased NAc core and shell c-Fos relative to animals that received saline, with animals receiving both E2 microinjections and systemic cocaine expressing the highest activation in the caudal NAc, compared to rats receiving aCSF microinjections and systemic cocaine (p = 0.05, d = 0.70). Cocaine 31-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 29888304-10 2018 However, high Fos expression was seen after cocaine in both reward- and stress-related brain regions of LBN rats, including nucleus accumbens core, central amygdala, and lateral habenula. Cocaine 44-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 29170000-7 2018 This severe impact of a tonically impaired PPT cholinergic innervation was evidenced as the cholinergic interneuronal loss of the caudate putamen and as a suppressed c-Fos expression after stimulation by d-AMPH. Dextroamphetamine 204-210 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-171 29080930-7 2018 Immunohistochemical staining revealed a significant reduction in the ratio of c-Fos-positive GABAergic neurons in the parafacial zone (PZ) beginning from day 7 after STZ-icv. Streptozocin 166-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 28120151-5 2018 Although unilateral intrahippocampal injection of either the A1R agonist CHA or the A2AR agonist CGS21680 increased pERK1/2, only CHA mitigated histopathological and behavioral deficits along with reducing glutamate, microglial activation, c-Fos, TNF-alpha, iNOS, TBARS, cytochrome c and caspase-3 and elevating Nrf2 and IL-10 levels in IR rats. 2-(4-(2-carboxyethyl)phenethylamino)-5'-N-ethylcarboxamidoadenosine 97-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 240-245 30019701-0 2018 Fos immunoreactivity in the intermediolateral nucleus induced by tendon vibration of the m. triceps surae in rats pretreated with a nitric oxide blocker or precursor. Nitric Oxide 132-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 28866130-11 2018 CORT118335 upregulated c-Fos expression in the paraventricular nucleus of the hypothalamus. CORT118335 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 28866130-12 2018 Imipramine decreased c-Fos in the basolateral amygdala and hippocampus (CA1 and CA3), but increased c-Fos in the central amygdala. Imipramine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 28866130-12 2018 Imipramine decreased c-Fos in the basolateral amygdala and hippocampus (CA1 and CA3), but increased c-Fos in the central amygdala. Imipramine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 28695319-0 2018 Acute Impact of Selected Pyridoindole Derivatives on Fos Expression in Different Structures of the Rat Brain. 1H-pyrrolo[2,3-f]quinoline 25-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 28695319-6 2018 In all the three groups of animals, the upregulation of PDs-induced Fos expression only in 2 of 11 brain areas was investigated, namely, in the hypothalamic paraventricular nucleus (PVN) and the central amygdaloid nucleus (CeA). 3-{1-[3-(Dimethylamino)propyl]-2-Methyl-1h-Indol-3-Yl}-4-(2-Methyl-1h-Indol-3-Yl)-1h-Pyrrole-2,5-Dione 56-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 28695319-8 2018 Counterstaining of the Fos-labeled CeA-containing sections with VIP antibody revealed that the Fos expression stimulated by the PDs was upregulated in all the CeA subdivisions (lateral, ventral, capsular), except the medial one. 3-{1-[3-(Dimethylamino)propyl]-2-Methyl-1h-Indol-3-Yl}-4-(2-Methyl-1h-Indol-3-Yl)-1h-Pyrrole-2,5-Dione 128-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 28695319-8 2018 Counterstaining of the Fos-labeled CeA-containing sections with VIP antibody revealed that the Fos expression stimulated by the PDs was upregulated in all the CeA subdivisions (lateral, ventral, capsular), except the medial one. 3-{1-[3-(Dimethylamino)propyl]-2-Methyl-1h-Indol-3-Yl}-4-(2-Methyl-1h-Indol-3-Yl)-1h-Pyrrole-2,5-Dione 128-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 30019701-7 2018 These results indicate that decreased nitric oxide release after injection of 7-NI was accompanied by a potentiation of the early c-fos gene expression induced by muscle proprioceptive activity within the thoracolumbar region of the rat spinal cord. Nitric Oxide 38-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 30019701-7 2018 These results indicate that decreased nitric oxide release after injection of 7-NI was accompanied by a potentiation of the early c-fos gene expression induced by muscle proprioceptive activity within the thoracolumbar region of the rat spinal cord. 7-nitroindazole 78-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 30157705-9 2018 suppressed formalin-evoked inflammatory pain in rats and reduced Fos protein-like immunoreactivity in the lumbar spinal dorsal horn. Formaldehyde 11-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 28464341-4 2018 Novel open field exposure in both CC and MC resulted in a marked increase in Fos expression in the anterior and posterior parts of the basolateral amygdaloid nucleus, basomedial nucleus, and medial nucleus, whereas these increases in expression were not observed in CR. Methylcholanthrene 41-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-80 29139213-6 2018 After 10 days of cocaine self-administration, both the density and intensity of c-fos-positive cells were significantly increased in LHbL, but not LHbM, while after 60 days, an increased density (but not intensity) of labeled neurons in both LHbL and LHbM was observed. Cocaine 17-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 29139213-6 2018 After 10 days of cocaine self-administration, both the density and intensity of c-fos-positive cells were significantly increased in LHbL, but not LHbM, while after 60 days, an increased density (but not intensity) of labeled neurons in both LHbL and LHbM was observed. lhbl 133-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 29222058-0 2018 Differential expression of the immediate early genes c-Fos, Arc, Egr-1, and Npas4 during long-term memory formation in the context preexposure facilitation effect (CPFE). cpfe 164-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 29088985-6 2018 Furthermore, immunofluorescence was used to investigate the influence of GM on the c-fos expression in the hippocampus. Gentamicins 73-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 29088985-11 2018 Additionally, the expression of c-fos was significantly decreased in the ipsilateral hippocampus of KA-injected rats treated with GM compared with KA-injected rats treated with saline. Gentamicins 130-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 29037790-9 2018 Intra-NAc administration of dexamethasone (1.0 and 3.0mug/site) facilitated the active nose-poke responses, which was accompanied by the upregulation of D1 receptor and c-Fos in the NAc. Dexamethasone 28-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-174 29054430-0 2018 NMDA receptor dependent changes in c-fos and p-CREB signaling following extinction and reinstatement of morphine place preference. Morphine 104-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 29054430-7 2018 Therefore, it can be assumed that consolidation and reconsolidation of morphine memory via intra-PFC, -NAc and -HIP NMDA glutamate receptors are in accordance with changes in p-CREB/CREB ratio and c-fos levels. Morphine 71-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 197-202 30270724-5 2018 Although the single-sided formalin injection caused a significant bilateral increase in c-Fos-expressing neurons in the lateral parabrachial nucleus with slight projection-side dependence, the increase in the amplitude of postsynaptic excitatory currents and the number of c-Fos-expressing neurons in the central amygdala occurred predominantly on the right side regardless of the side of the inflammation. Formaldehyde 26-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 30270724-5 2018 Although the single-sided formalin injection caused a significant bilateral increase in c-Fos-expressing neurons in the lateral parabrachial nucleus with slight projection-side dependence, the increase in the amplitude of postsynaptic excitatory currents and the number of c-Fos-expressing neurons in the central amygdala occurred predominantly on the right side regardless of the side of the inflammation. Formaldehyde 26-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 273-278 29311809-3 2017 Our previous work has shown that transcriptional activation of c-Fos and Egr-1 in the hippocampus requires formation of a dual histone mark within their promoter regions, the phosphorylation of serine 10 and acetylation of lysine 9/14 of histone H3. Serine 194-200 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 29311809-3 2017 Our previous work has shown that transcriptional activation of c-Fos and Egr-1 in the hippocampus requires formation of a dual histone mark within their promoter regions, the phosphorylation of serine 10 and acetylation of lysine 9/14 of histone H3. Lysine 223-229 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 29311809-4 2017 In the present study, using chromatin immuno-precipitation (ChIP), we found that an increase in the formation of H3K9ac-S10p occurs within the c-Fos and Egr-1 promoters after FS stress in vivo and that these histone modifications were located to promoter regions containing cAMP Responsive Elements (CREs), but not in neighboring regions containing only Serum Responsive Elements (SREs). Cyclic AMP 274-278 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-148 28951234-8 2017 Moreover, we observed significantly more c-Fos and nesfatin-1 ir double-labeled cells in ABA rats compared to RF, AL and AC in the supraoptic nucleus (p<0.05) and compared to AL and AC in the paraventricular nucleus, arcuate nucleus, dorsomedial hypothalamic nucleus, dorsal raphe nucleus and the rostral raphe pallidus (p<0.05). alisol B 23-acetate 89-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 29325386-16 2017 MK-801 or PD98059 intervention further promote the Nissl body volume reduced, neurons karyopyknosis, the apoptosis of visual cortical neurons and Caspase-3 expression, and restrain the expression of NGF and c-FOS. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 207-212 29325386-16 2017 MK-801 or PD98059 intervention further promote the Nissl body volume reduced, neurons karyopyknosis, the apoptosis of visual cortical neurons and Caspase-3 expression, and restrain the expression of NGF and c-FOS. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 10-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 207-212 28762073-8 2017 cFos expression significantly increased in the dorsomedial striatum and major subregions of the medial prefrontal cortex (mPFC) in the morphine group. Morphine 135-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-4 29348799-6 2017 We showed here that an intraperitoneal injection of ethanol (0.25 g/kg), resulting in a blood ethanol concentration of 5.6 mM, significantly increased the number of cFos immunoreactive (IR) neurons in the LHb. Ethanol 52-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-169 29348799-6 2017 We showed here that an intraperitoneal injection of ethanol (0.25 g/kg), resulting in a blood ethanol concentration of 5.6 mM, significantly increased the number of cFos immunoreactive (IR) neurons in the LHb. Ethanol 94-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-169 29348799-6 2017 We showed here that an intraperitoneal injection of ethanol (0.25 g/kg), resulting in a blood ethanol concentration of 5.6 mM, significantly increased the number of cFos immunoreactive (IR) neurons in the LHb. 5-[[[(2~{r},3~{s},4~{r},5~{r})-5-(6-Aminopurin-9-Yl)-3,4-Bis(Oxidanyl)oxolan-2-Yl]methylamino]methyl]-4-Azanyl-1-(Methoxymethyl)pyrimidin-2-One 205-208 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-169 29348799-7 2017 Most of the ethanol-activated cFos-IR LHb neurons expressed vGluT2 (vesicular glutamate transporters 2, a marker of a glutamatergic phenotype). Ethanol 12-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-34 29348799-7 2017 Most of the ethanol-activated cFos-IR LHb neurons expressed vGluT2 (vesicular glutamate transporters 2, a marker of a glutamatergic phenotype). 5-[[[(2~{r},3~{s},4~{r},5~{r})-5-(6-Aminopurin-9-Yl)-3,4-Bis(Oxidanyl)oxolan-2-Yl]methylamino]methyl]-4-Azanyl-1-(Methoxymethyl)pyrimidin-2-One 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-34 29098175-4 2017 We first showed that approximately half of the pubertally born AVPV cells are activated by estradiol plus progesterone (P) treatment, as demonstrated by Fos expression, and that approximately 10% of pubertally born AVPV cells express estrogen receptor alpha (ERalpha). Estradiol 91-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-156 29098175-4 2017 We first showed that approximately half of the pubertally born AVPV cells are activated by estradiol plus progesterone (P) treatment, as demonstrated by Fos expression, and that approximately 10% of pubertally born AVPV cells express estrogen receptor alpha (ERalpha). Progesterone 106-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-156 29089891-8 2017 Ethanol-associated context induced the reinstatement of ethanol seeking and increased the expression of Fos in the prelimbic cortex. Ethanol 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 28739394-8 2017 PCBs decreased the protein levels of FSHR, AR, ABP, ERalpha & beta, transferrin, claudin-11, occludin, E-cadherin, connexin-43, c-fos, c-jun, SF1, USF1 & 2 whereas inhibin-beta protein level was found to be increased in Sertoli cells. Polychlorinated Biphenyls 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 28787345-9 2017 To determine whether 5 daily injections of amitriptyline activated noradrenergic neurons in the locus coeruleus (LC) and spinal cord with or without DSP-4 treatment, we performed immunohistochemistry using antibodies for c-Fos and dopamine beta-hydroxylase (DbetaH). Amitriptyline 43-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 221-226 28787345-13 2017 Additionally, 5 daily injections of amitriptyline increased the ratio of c-Fos-immunoreactive (IR) cells in noradrenergic LC neurons in SNL rats with or without DSP-4 pretreatment (P < .001, respectively). Amitriptyline 36-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 28726252-9 2017 Furthermore, yohimbine-induced reinstatement of alcohol seeking increased Fos activation in CeA corticotrophin-releasing factor, dynorphin and GABA neurons compared with naive and vehicle controls. Yohimbine 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 28726252-9 2017 Furthermore, yohimbine-induced reinstatement of alcohol seeking increased Fos activation in CeA corticotrophin-releasing factor, dynorphin and GABA neurons compared with naive and vehicle controls. Alcohols 48-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 28726252-9 2017 Furthermore, yohimbine-induced reinstatement of alcohol seeking increased Fos activation in CeA corticotrophin-releasing factor, dynorphin and GABA neurons compared with naive and vehicle controls. gamma-Aminobutyric Acid 143-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 27048408-8 2017 Furthermore, both Cvb-D, captopril and SB203580 reduced mRNA expression of p38alpha, p38beta, c-fos, and c-jun mRNA, and Cvb-D had a stronger effect (P<0.01). Captopril 25-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 27048408-8 2017 Furthermore, both Cvb-D, captopril and SB203580 reduced mRNA expression of p38alpha, p38beta, c-fos, and c-jun mRNA, and Cvb-D had a stronger effect (P<0.01). SB 203580 39-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 28799666-0 2017 Effect of resveratrol on c-fos expression of rat trigeminal spinal nucleus caudalis and C1 dorsal horn neurons following mustard oil-induced acute inflammation. Resveratrol 10-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 28799666-0 2017 Effect of resveratrol on c-fos expression of rat trigeminal spinal nucleus caudalis and C1 dorsal horn neurons following mustard oil-induced acute inflammation. mustard oil 121-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 28799666-2 2017 The aim of the present study was to investigate whether pretreatment with resveratrol attenuates acute inflammation-induced sensitization of nociceptive processing in rat spinal trigeminal nucleus caudalis (SpVc) and upper cervical (C1) dorsal horn neurons, via c-fos immunoreactivity. Resveratrol 74-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 262-267 28931335-4 2017 Intraplantar injection of H2O2 (5, 10 and 20 micromoles/paw) induced a significant thermal hyperalgesia in the hind paw, confirmed by increased c-Fos activity in dorsal horn of spinal cord. Hydrogen Peroxide 26-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-149 28977524-10 2017 By contrast, forced swimming test-induced c-FOS immunoreactivity was reduced in dorsal dentate gyrus and ventral CA1 following treatment with TRIM or ZL006. 4-((3,5-dichloro-2-hydroxybenzyl)amino)-2-hydroxybenzoic acid 150-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 28977524-13 2017 Conclusions: This study identified a pattern of enhanced and reduced forced swimming test-related c-FOS immunoreactivity indicative of a regulated network where inhibition of nitric oxide coupled to the N-methyl-D-aspartic acid receptor leads to activation of the lateral septum, periaqueductal gray, and paraventricular nucleus of the hypothalamus with concomitant inhibition of the hippocampus. Nitric Oxide 175-187 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 28977525-0 2017 Oxytocin Reduces Cocaine Cued Fos Activation in a Regionally Specific Manner. Cocaine 17-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 28977525-4 2017 Here, we studied Fos expression following cocaine-cued reinstatement in both male and female rats. Cocaine 42-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 28842814-11 2017 Activation of PKCgamma by PMA aggravated allodynia and increased the expression of CGRP and c-Fos. Tetradecanoylphorbol Acetate 26-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 29088985-11 2018 Additionally, the expression of c-fos was significantly decreased in the ipsilateral hippocampus of KA-injected rats treated with GM compared with KA-injected rats treated with saline. Sodium Chloride 177-183 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 28687229-0 2017 The phosphodiesterase 10A selective inhibitor, TAK-063, induces c-Fos expression in both direct and indirect pathway medium spiny neurons and sub-regions of the medial prefrontal cortex in rats. 1-(2-fluoro-4-(1H-pyrazol-1-yl)phenyl)-5-methoxy-3-(1-phenyl-1H-pyrazol-5-yl)pyridazin-4(1H)-one 47-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 28687229-4 2017 TAK-063 at 0.3 and 3mg/kg induced a dose-dependent increase in the number of c-Fos immunoreactive cells in the striatal complex. 1-(2-fluoro-4-(1H-pyrazol-1-yl)phenyl)-5-methoxy-3-(1-phenyl-1H-pyrazol-5-yl)pyridazin-4(1H)-one 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 28687229-5 2017 Furthermore, TAK-063 increased the number of MSNs expressing c-fos mRNA in both the D1 receptor-expressing direct pathway and D2 receptor-expressing indirect pathway of the striatal complex. 1-(2-fluoro-4-(1H-pyrazol-1-yl)phenyl)-5-methoxy-3-(1-phenyl-1H-pyrazol-5-yl)pyridazin-4(1H)-one 13-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 28687229-6 2017 TAK-063 (0.3 and 3mg/kg) induced c-Fos expression in the anterior cingulate cortex (ACC) and prelimbic cortex (PrL), but not the infralimbic, dorsal peduncular, primary motor or anterior insular cortices. 1-(2-fluoro-4-(1H-pyrazol-1-yl)phenyl)-5-methoxy-3-(1-phenyl-1H-pyrazol-5-yl)pyridazin-4(1H)-one 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 27794548-9 2017 At one week following RTX, decreased c-Fos and primary afferent neuropeptide immunoreactivities were observed in the dorsal horn, while plantar burn pathology was unaltered. resiniferatoxin 22-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 27730727-5 2017 In addition, treatment with another selective TRPV1 antagonist, AMG9810, not only significantly prevented morphine self-administration but also prevented morphine-induced c-fos expression in the nucleus accumbens. Morphine 154-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-176 28322236-5 2017 Systemic clozapine-n-oxide (CNO; 0.3 mg/kg) successfully recruited a large proportion of the VTA-NAc dopaminergic projections, with activity evidenced by colocalization with elevated levels of Fos protein. clozapine N-oxide 9-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-196 28322236-5 2017 Systemic clozapine-n-oxide (CNO; 0.3 mg/kg) successfully recruited a large proportion of the VTA-NAc dopaminergic projections, with activity evidenced by colocalization with elevated levels of Fos protein. clozapine N-oxide 28-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-196 29104261-0 2017 Neurochemical Changes and c-Fos Mapping in the Brain after Carisbamate Treatment of Rats Subjected to Lithium-Pilocarpine-Induced Status Epilepticus. S-2-O-carbamoyl-1-o-chlorophenyl-ethanol 59-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 28966579-7 2017 Inhibition of the MS with muscimol pre-treatment attenuated both carbachol-evoked c-Fos-ir and theta activation. Muscimol 26-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 28951360-5 2017 Compared with BK + glutamate group, intrathecal administration of methysergide significantly attenuated the inhibitory effect of chemical stimulation of the LRN on intrapericardial BK-induced EMG and increased c-Fos expression in the spinal dorsal horn (P<0.05). Methysergide 66-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 210-215 28966579-7 2017 Inhibition of the MS with muscimol pre-treatment attenuated both carbachol-evoked c-Fos-ir and theta activation. Carbachol 65-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 28884307-7 2017 This effect was associated to a significant inhibition of nitroglycerin-induced increase in c-Fos, TRPA1 and neuropeptides mRNA levels in medulla-pons area, in the cervical spinal cord and in the trigeminal ganglion. Nitroglycerin 58-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 28093219-11 2017 Imipramine decreased c-Fos expression in the basolateral amygdala and CA1 and CA3 divisions of the hippocampus. Imipramine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 28697452-5 2017 Fluoride poisoning induced severe cell injuries, such as decreased PC12 cell activity, enhanced cell apoptosis, high c-fos, CAMKII, and Bax mRNA expression levels. Fluorides 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 28954467-6 2017 METHODS: In the present study, the effects of tamsulosin on the expression of c-Fos, nerve growth factor (NGF), and nicotinamide adenine dinucleotide phosphate-diaphorase (NADPH-d) in the afferent micturition areas, including the pontine micturition center (PMC), the ventrolateral periaqueductal gray matter (vlPAG), and the spinal cord (L5), of rats with an SCI were investigated. Tamsulosin 46-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 28912720-7 2017 Elevated Fos expression in the PVN was also present at 24 h (by 73 +- 11%) following Ang II compared to control saline injections, confirming persistent activation of PVN. Sodium Chloride 112-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 28634074-5 2017 Consistent with this increase, c-fos expression was induced in a KCl concentration-dependent manner. Potassium Chloride 65-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 28825095-0 2017 [Effect of methylphenidate on c-Fos expression in parvalbumin interneurons of juvenile rat frontal cortex]. Methylphenidate 11-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 28474155-9 2017 Likewise, NA-2 and indomethacin treatment also significantly suppressed the mRNA expression of RANKL, RANK, c-fos, c-jun, NF-kappaB (p < 0.0001) and Akt (p < 0.01) and protein expression of iNOS, pAkt and c-Fos (p < 0.0001) compared to the arthritic control group. Indomethacin 19-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 28474155-9 2017 Likewise, NA-2 and indomethacin treatment also significantly suppressed the mRNA expression of RANKL, RANK, c-fos, c-jun, NF-kappaB (p < 0.0001) and Akt (p < 0.01) and protein expression of iNOS, pAkt and c-Fos (p < 0.0001) compared to the arthritic control group. Indomethacin 19-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 211-216 28720013-8 2017 Our results show that CP94253 (3, 10 mg/kg) produced a dose-dependent potentiation of methylphenidate (5 mg/kg)-induced expression of Zif268 and c- Fos. CP 94253 22-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-151 28527956-0 2017 Comparison of the induction of c-fos-eGFP and Fos protein in the rat spinal cord and hypothalamus resulting from subcutaneous capsaicin or formalin injection. Capsaicin 126-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 28527956-0 2017 Comparison of the induction of c-fos-eGFP and Fos protein in the rat spinal cord and hypothalamus resulting from subcutaneous capsaicin or formalin injection. Formaldehyde 139-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 28527956-0 2017 Comparison of the induction of c-fos-eGFP and Fos protein in the rat spinal cord and hypothalamus resulting from subcutaneous capsaicin or formalin injection. Formaldehyde 139-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 28527956-7 2017 Following capsaicin or formalin treatment, eGFP was maximally expressed at 6h in the spinal cord and 3h in the PVN and SON, whereas, Fos-LI was maximally expressed at 1.5h in all the regions we analyzed. Formaldehyde 23-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-136 28527956-8 2017 Induction of eGFP in the OXT neurons was observed after capsaicin or formalin treatment, while Fos-LI in the OXT neurons was observed only after formalin treatment. Formaldehyde 145-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 28730839-0 2017 TRPV1 receptors contribute to paclitaxel-induced c-Fos expression in spinal cord dorsal horn neurons. Paclitaxel 30-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 28730839-6 2017 PAC treatment induced significant upregulation of c-Fos nuclear expression in superficial dorsal horn neurons that was diminished by TRPV1 receptor antagonists pre-incubation. Paclitaxel 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 28190104-2 2017 The immediate early gene c-fos can be used to gain insight into the activity of MCs in vivo, because c-fos protein expression reflects increased neuronal activity. mcs 80-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 28720013-8 2017 Our results show that CP94253 (3, 10 mg/kg) produced a dose-dependent potentiation of methylphenidate (5 mg/kg)-induced expression of Zif268 and c- Fos. Methylphenidate 86-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-151 28190104-2 2017 The immediate early gene c-fos can be used to gain insight into the activity of MCs in vivo, because c-fos protein expression reflects increased neuronal activity. mcs 80-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 28190104-4 2017 In this study, we hypothesized that restraint stress would alter c-fos-ir, because MCs express glucocorticoid type 2 receptors and the DG is considered to be involved in behaviors related to stress or anxiety. mcs 83-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 28190104-5 2017 We show that acute restraint using a transparent nose cone for just 10 min led to reduced c-fos-ir in ventral MCs compared to control rats. mcs 110-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 27535568-10 2017 In the 0.75 g/kg L-histidine group, a significant increase in the number of Fos-ir cells was detected only in the NTS. Histidine 17-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 28295336-3 2017 Here we determine the neurochemistry (using in situ hybridization) of catecholaminergic and noncatecholaminergic neurons which express c-Fos immunoreactivity throughout the rostrocaudal extent of the ventrolateral medulla, in Sprague Dawley rats treated with hydralazine or saline. Hydralazine 259-270 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 28295336-3 2017 Here we determine the neurochemistry (using in situ hybridization) of catecholaminergic and noncatecholaminergic neurons which express c-Fos immunoreactivity throughout the rostrocaudal extent of the ventrolateral medulla, in Sprague Dawley rats treated with hydralazine or saline. Sodium Chloride 274-280 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 28343071-11 2017 Meanwhile, immunohistochemistry results showed that NTG treatment significantly activated c-Fos neurons while BAI treatment inhibited the expression of c-Fos. Nitroglycerin 52-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 28580417-6 2017 Post-synaptically, pHFD animals display an increase in NAc D2 receptors and c-Fos expression after amphetamine injection. Amphetamine 99-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 28274653-7 2017 The results showed that kainic acid infusion into the lwDR increased Fos protein immunostaining in brain structures deeply involved in panic-like defensive behaviors, such as the periaqueductal gray and hypothalamus, but not the amygdala. Kainic Acid 24-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 28259933-10 2017 Furthermore, the combination of alfentanil and propofol treatments may produce synergistically antinociceptive effects by inhibiting the phosphorylation of ERK1/2 and decreasing the expression of c-fos in the spinal cord. Alfentanil 32-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 196-201 28285942-0 2017 Small molecule inhibitors of PSD95-nNOS protein-protein interactions suppress formalin-evoked Fos protein expression and nociceptive behavior in rats. Formaldehyde 78-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 28285942-6 2017 We examined the impact of small molecule PSD95-nNOS protein-protein interaction inhibitors (ZL006, IC87201) on both nociceptive behavior and formalin-evoked Fos protein expression within the lumbar spinal cord of rats. Formaldehyde 141-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-160 28285942-8 2017 IC87201 and ZL006, but not ZL007, suppressed phase 2 of formalin-evoked pain behavior and decreased the number of formalin-induced Fos-like immunoreactive cells in spinal dorsal horn regions associated with nociceptive processing. Formaldehyde 114-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-134 28469203-5 2017 Furthermore, the protein levels of phosphorylated cAMP response element binding protein (p-CREB) and c-fos were also increased, which were blocked by co-injecting ERK inhibitor FR180204. FR 180204 177-185 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 28212941-0 2017 Injections of the of the alpha1-adrenoceptor antagonist prazosin into the median raphe nucleus increase food intake and Fos expression in orexin neurons of free-feeding rats. Prazosin 56-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-123 28212941-4 2017 For this purpose, we examined the induction of c-Fos immunoreactivity in forebrain structures following injections of the alpha1-adrenoceptor antagonist prazosin into the MnR of free-feeding rats. Prazosin 153-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 28179107-4 2017 Pretreatment with the nonselective COX inhibitor, indomethacin, either icv (10mug in 5mul) or iv (1mg/kg) significantly reduced restraint-induced Fos expression in the paraventricular hypothalamic nucleus (PVH) by 45%, relative to vehicle-injected controls. Indomethacin 50-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-149 28179107-4 2017 Pretreatment with the nonselective COX inhibitor, indomethacin, either icv (10mug in 5mul) or iv (1mg/kg) significantly reduced restraint-induced Fos expression in the paraventricular hypothalamic nucleus (PVH) by 45%, relative to vehicle-injected controls. icv 71-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-149 28609288-0 2017 Eff ect of a single asenapine treatment on Fos expression in the brain catecholamine-synthesizing neurons: impact of a chronic mild stress preconditioning. asenapine 20-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 28609288-0 2017 Eff ect of a single asenapine treatment on Fos expression in the brain catecholamine-synthesizing neurons: impact of a chronic mild stress preconditioning. Catecholamines 71-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 28473751-5 2017 Also, the nuclear expression of c-Fos, a marker for neuronal activation, related to water-consumption (SFO and MnPO) was inhibited. Water 84-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 28320839-10 2017 Caffeine treatment in midnight triggered c-Fos expression in dorsal SCN. Caffeine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 28320839-11 2017 Both sleep deprivation and caffeine treatment potentiated light-induced c-Fos expression in calbindin-containing cells of the ventral SCN in early and late night. Caffeine 27-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 28374767-7 2017 GPR91 siRNA transduction and the ERK1/2 inhibitor U0126 inhibited the increases in C/EBP beta, C/EBP delta, c-Fos and HIF-1alpha. U 0126 50-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 28285942-9 2017 MK-801 suppressed Fos protein expression in both dorsal and ventral horns. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 28323090-0 2017 c-Fos activity in the insular cortex, nucleus accumbens and basolateral amygdala following the intraperitoneal injection of saccharin and lithium chloride. Saccharin 124-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 28323090-0 2017 c-Fos activity in the insular cortex, nucleus accumbens and basolateral amygdala following the intraperitoneal injection of saccharin and lithium chloride. Lithium Chloride 138-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 28323090-1 2017 This study examined c-Fos expression in selected brain areas consequent to intraperitoneal (IP) administration of saccharin and lithium chloride. Saccharin 114-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 28323090-1 2017 This study examined c-Fos expression in selected brain areas consequent to intraperitoneal (IP) administration of saccharin and lithium chloride. Lithium Chloride 128-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 28221079-8 2017 We also assessed Fos expression in LH orexin and GABA neurons. gamma-Aminobutyric Acid 49-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 28221079-10 2017 Relapse induced by the appetitive CS was associated with increased Fos expression in LH, caudal basolateral amygdala (BLA), and medial amygdala (MeA). Cesium 34-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 27295092-8 2017 The c-fos protein levels of the C8-T5 lateral horn neurons and the blood catecholamine levels were increased in the nicotine group, but the cervical vagal activity was not changed. Nicotine 116-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 28092166-6 2017 Pretreatment of the I-R animals with metformin increased phosphorylated cyclic-AMP response element-binding protein (pCREB) and c-fos levels compared to the I-R group (P < 0.001 for both). Metformin 37-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-133 28259933-10 2017 Furthermore, the combination of alfentanil and propofol treatments may produce synergistically antinociceptive effects by inhibiting the phosphorylation of ERK1/2 and decreasing the expression of c-fos in the spinal cord. Propofol 47-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 196-201 28030475-3 2017 Nuclear mGluR5 is a functional receptor that binds glutamate entering the cell through the neuronal glutamate transporter (GT) EAAT3 and activates transcription factor c-fos, whereas plasma membrane mGluR5 is responsible for c-jun activation. Glutamic Acid 51-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-173 28087833-4 2017 Moreover, DPAA induces up-regulation of oxidative stress-counteracting proteins, activation of CREB phosphorylation, increased protein expression of c-Jun and c-Fos, and aberrant secretion of brain-active cytokines (MCP-1, adrenomedullin, FGF2, CXCL1, and IL-6). diphenylarsinic acid 10-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-164 28131625-8 2017 We then showed that CUS compromised excitatory afferent activation of the mPFC following pharmacological stimulation of the mediodorsal thalamus (MDT), indicated by a reduced induction of c-fos expression. cus 20-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 188-193 27468916-5 2017 In rats, we assessed Fos response to lithium chloride (LiCl), beta-carboline, naloxone, lipopolysaccharide (LPS), inflammatory pain, neuropathic pain, foot-shock, restraint stress, forced swimming, predator odor, and opiate withdrawal. Lithium Chloride 37-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 29026496-1 2017 OBJECTIVES: To investigate the effects of Nonylphenol (NP) in pups from dams exposed during gestational and lactational periods on immediate early genes (c-jun, c-fos) in hippocampus and the learning and memory of F1 rats. nonylphenol 42-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-166 28109977-4 2017 In normal control animals, intraoral application of 1mM quinine caused increased numbers of c-Fos-immunoreactive (c-Fos-IR) neurons in the external lateral subnucleus and external medial subnucleus of the PBN (elPBN and emPBN, respectively) compared with application of distilled water. Quinine 56-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 28109977-4 2017 In normal control animals, intraoral application of 1mM quinine caused increased numbers of c-Fos-immunoreactive (c-Fos-IR) neurons in the external lateral subnucleus and external medial subnucleus of the PBN (elPBN and emPBN, respectively) compared with application of distilled water. Quinine 56-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 29931935-9 2017 CONCLUSIONS: Elevated proliferation and expression of PKCalpha, c-fos, Bcl-2 was observed in rat PASMCs in hypoxia, and while the apoptosis rate had no significant change. pasmcs 97-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 28089665-11 2017 Lastly, formalin-induced cFos expression and the effects of systemic morphine were examined in the superficial dorsal horn of the spinal cord. Formaldehyde 8-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-29 28089665-12 2017 Intraplantar formalin produced robust expression of cFos; however, morphine did not attenuate the cFos expression. Formaldehyde 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-56 27053349-2 2017 In addition, several lines of study have indicated that cAMP response element-binding protein (CREB) and c-fos have important role in morphine-induced conditioned place preference (CPP) induced by drugs of abuse, such as morphine, cocaine, nicotine, and alcohol. Morphine 134-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 27053349-2 2017 In addition, several lines of study have indicated that cAMP response element-binding protein (CREB) and c-fos have important role in morphine-induced conditioned place preference (CPP) induced by drugs of abuse, such as morphine, cocaine, nicotine, and alcohol. Morphine 221-229 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 27053349-2 2017 In addition, several lines of study have indicated that cAMP response element-binding protein (CREB) and c-fos have important role in morphine-induced conditioned place preference (CPP) induced by drugs of abuse, such as morphine, cocaine, nicotine, and alcohol. Cocaine 231-238 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 27053349-2 2017 In addition, several lines of study have indicated that cAMP response element-binding protein (CREB) and c-fos have important role in morphine-induced conditioned place preference (CPP) induced by drugs of abuse, such as morphine, cocaine, nicotine, and alcohol. Nicotine 240-248 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 27053349-2 2017 In addition, several lines of study have indicated that cAMP response element-binding protein (CREB) and c-fos have important role in morphine-induced conditioned place preference (CPP) induced by drugs of abuse, such as morphine, cocaine, nicotine, and alcohol. Alcohols 254-261 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 28263512-7 2017 RESULTS: In metoclopramide groups brain stem c-fos expression was significantly lower than in the CSD side of the saline group (P = 0.002). Metoclopramide 12-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 28263512-8 2017 In the raclopride group, ipsilateral brain stem c-fos expression was also lower than in the saline group (P = 0.002). Raclopride 7-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 27856260-8 2017 Furthermore, Fos protein-labelled neurons were observed in the dPAG after the stimulation of the dmVMH with 9nmol of NMDA. dpag 63-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 27856260-8 2017 Furthermore, Fos protein-labelled neurons were observed in the dPAG after the stimulation of the dmVMH with 9nmol of NMDA. dmvmh 97-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 27856260-8 2017 Furthermore, Fos protein-labelled neurons were observed in the dPAG after the stimulation of the dmVMH with 9nmol of NMDA. N-Methylaspartate 117-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 27856260-9 2017 Additionally, when the anterograde neurotracer biotinylated dextran amine (BDA) was deposited in the dmVMH subsequent stimulation of the dmVMH produced BDA-labelled neural fibres with terminal boutons surrounding Fos-labelled neurons in the dPAG, suggesting synaptic contacts between dmVMH and dPAG neurons for eliciting panic-like behavioural responses. dextran amine 60-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-216 27856260-9 2017 Additionally, when the anterograde neurotracer biotinylated dextran amine (BDA) was deposited in the dmVMH subsequent stimulation of the dmVMH produced BDA-labelled neural fibres with terminal boutons surrounding Fos-labelled neurons in the dPAG, suggesting synaptic contacts between dmVMH and dPAG neurons for eliciting panic-like behavioural responses. biotinylated dextran amine 75-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-216 27856260-9 2017 Additionally, when the anterograde neurotracer biotinylated dextran amine (BDA) was deposited in the dmVMH subsequent stimulation of the dmVMH produced BDA-labelled neural fibres with terminal boutons surrounding Fos-labelled neurons in the dPAG, suggesting synaptic contacts between dmVMH and dPAG neurons for eliciting panic-like behavioural responses. dmvmh 137-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-216 27856260-9 2017 Additionally, when the anterograde neurotracer biotinylated dextran amine (BDA) was deposited in the dmVMH subsequent stimulation of the dmVMH produced BDA-labelled neural fibres with terminal boutons surrounding Fos-labelled neurons in the dPAG, suggesting synaptic contacts between dmVMH and dPAG neurons for eliciting panic-like behavioural responses. dmvmh 137-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-216 28232801-7 2017 Topographical analysis of Fos protein expression, a biological marker of neural excitation, revealed that a convulsive dose (4 mg/kg) of nicotine region-specifically activated neurons in the piriform cortex, amygdala, medial habenula, paratenial thalamus, anterior hypothalamus and solitary nucleus among 48 brain regions examined, and this was also suppressed by MEC. Nicotine 137-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 27468916-7 2017 Naloxone-precipitated opiate withdrawal induced Fos in mu-opioid receptor-positive (15%) and -negative (85%) tVTA cells, suggesting the presence of both direct and indirect mechanisms in tVTA recruitment during withdrawal. Naloxone 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 27468916-7 2017 Naloxone-precipitated opiate withdrawal induced Fos in mu-opioid receptor-positive (15%) and -negative (85%) tVTA cells, suggesting the presence of both direct and indirect mechanisms in tVTA recruitment during withdrawal. Opiate Alkaloids 22-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 27468916-7 2017 Naloxone-precipitated opiate withdrawal induced Fos in mu-opioid receptor-positive (15%) and -negative (85%) tVTA cells, suggesting the presence of both direct and indirect mechanisms in tVTA recruitment during withdrawal. tvta 109-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 28123032-8 2017 The incubated response was associated with increased Fos expression in DMS but not in DLS; Fos was colabeled with both Drd1 and Drd2 DMS injections of SCH39166 or raclopride selectively decreased methamphetamine seeking after 21 abstinence days. ecopipam 151-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 28123032-8 2017 The incubated response was associated with increased Fos expression in DMS but not in DLS; Fos was colabeled with both Drd1 and Drd2 DMS injections of SCH39166 or raclopride selectively decreased methamphetamine seeking after 21 abstinence days. ecopipam 151-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-94 28123032-8 2017 The incubated response was associated with increased Fos expression in DMS but not in DLS; Fos was colabeled with both Drd1 and Drd2 DMS injections of SCH39166 or raclopride selectively decreased methamphetamine seeking after 21 abstinence days. Raclopride 163-173 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 28123032-8 2017 The incubated response was associated with increased Fos expression in DMS but not in DLS; Fos was colabeled with both Drd1 and Drd2 DMS injections of SCH39166 or raclopride selectively decreased methamphetamine seeking after 21 abstinence days. Raclopride 163-173 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-94 28123032-8 2017 The incubated response was associated with increased Fos expression in DMS but not in DLS; Fos was colabeled with both Drd1 and Drd2 DMS injections of SCH39166 or raclopride selectively decreased methamphetamine seeking after 21 abstinence days. Methamphetamine 196-211 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-94 28123032-9 2017 In Fos-lacZ transgenic rats, selective inactivation of relapse test-activated Fos neurons in DMS on abstinence day 18 decreased incubated methamphetamine seeking on day 21. Methamphetamine 138-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 3-6 28123032-9 2017 In Fos-lacZ transgenic rats, selective inactivation of relapse test-activated Fos neurons in DMS on abstinence day 18 decreased incubated methamphetamine seeking on day 21. Methamphetamine 138-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 27616342-9 2017 We further found that exposure to SPS significantly decreased c-Fos expression in the NAc core but not the shell after set-shifting behavior. Sodium phenolsulfonate 34-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 27572469-6 2016 The RVLM of PGE2-treated rats exhibited increases in c-Fos expression and extracellular signal-regulated kinase 1/2 and neuronal nitric oxide synthase phosphorylation along with oxidative stress, and PGE2 increased l-glutamate release in PC12 cells (surrogates of RVLM neurons). Dinoprostone 12-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 29093348-6 2017 Moreover, after the posttest, the number of cFos-positive mPFC neurons in rats that were conditioned with cocaine was significantly larger than that with saline. Cocaine 106-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-48 29093348-6 2017 Moreover, after the posttest, the number of cFos-positive mPFC neurons in rats that were conditioned with cocaine was significantly larger than that with saline. Sodium Chloride 154-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-48 27174774-7 2017 RESULTS: After administration of salicylate, c-fos protein expression increased significantly in the DCN, pVCN and IC when assayed by western blot. Salicylates 33-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 29363908-6 2017 Analysis of the pH3S10 and c-Fos expression levels in the group of D-amphetamine administered rats provided evidence of enhanced expression of these proteins in the regions of neurogenesis occurrence in rats. Dextroamphetamine 67-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 27703043-5 2017 After the second dose of amphetamine, the LR rats exhibited more c-Fos and GluN2B activation in layers II and III of the M1/M2 motor cortex area and prefrontal cortex (PRE, PRL, IL) and also presented with more GluN2B activation in the basal amygdala. Amphetamine 25-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 28969472-8 2017 Propofol also reduced the neuronal expression of c-Fos and p-ERK induced by formalin. Propofol 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 28969472-8 2017 Propofol also reduced the neuronal expression of c-Fos and p-ERK induced by formalin. Formaldehyde 76-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 27515792-7 2017 ED1 increased Fos expression in DH, LC, and DR, and DH Fos was decreased by systemic S-propranolol. Propranolol 85-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 28216890-8 2017 RESULTS: Extensive DNA damage in acute study and a significant increase in levels of p-MAPKs", c-fos, c-jun, and EGFR was observed in N-diethylnitrosamine (200 mg/kg bw) and ferric nitrilotriacetate (9 mg/kg bw) alone treated rats. Diethylnitrosamine 134-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 26410393-10 2016 CONCLUSION: Under the present experimental conditions, GOS/FOS mixture induced colonic positive effects, which increased Ca, P and Mg absorption. Phosphorus 126-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 26410393-10 2016 CONCLUSION: Under the present experimental conditions, GOS/FOS mixture induced colonic positive effects, which increased Ca, P and Mg absorption. Magnesium 132-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 27778243-11 2016 In addition, sulforaphane exposure effectively inhibited the expression of nNOS and c-Fos, reduced the number of nNOS and c-Fos immunoreactive neurons in TNC, and attenuated the tactile thresholds induced by NTG injection. sulforaphane 13-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 27778243-11 2016 In addition, sulforaphane exposure effectively inhibited the expression of nNOS and c-Fos, reduced the number of nNOS and c-Fos immunoreactive neurons in TNC, and attenuated the tactile thresholds induced by NTG injection. sulforaphane 13-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 26597361-3 2016 To elucidate mechanisms of action of fornix DBS with regard to memory restoration, we performed c-Fos immunohistochemistry in the hippocampus. fornix dbs 37-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 26646512-5 2017 Interestingly, this increase was associated with memory durability, since blocking c-Fos expression using specific antisense oligonucleotides (ASO) impaired long-lasting retention 7 days after training without affecting memory expression 2 days after training. Oligonucleotides 125-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 26646512-5 2017 Interestingly, this increase was associated with memory durability, since blocking c-Fos expression using specific antisense oligonucleotides (ASO) impaired long-lasting retention 7 days after training without affecting memory expression 2 days after training. Oligonucleotides, Antisense 143-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 28049513-9 2017 Chronic treatment with analgesics increased the CSD-evoked expression of Fos in the TNC and amygdala [18 +- 10.2 Fos-immunoreactive (IR) neurons per slide in the amygdala of rats treated with aspirin, 11 +- 5.4 IR neurons per slide in rats treated with acetaminophen, and 4 +- 3.7 IR neurons per slide in saline-treated control rats, P < 0.001]. Aspirin 192-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-76 28049513-9 2017 Chronic treatment with analgesics increased the CSD-evoked expression of Fos in the TNC and amygdala [18 +- 10.2 Fos-immunoreactive (IR) neurons per slide in the amygdala of rats treated with aspirin, 11 +- 5.4 IR neurons per slide in rats treated with acetaminophen, and 4 +- 3.7 IR neurons per slide in saline-treated control rats, P < 0.001]. Acetaminophen 253-266 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-76 28049513-9 2017 Chronic treatment with analgesics increased the CSD-evoked expression of Fos in the TNC and amygdala [18 +- 10.2 Fos-immunoreactive (IR) neurons per slide in the amygdala of rats treated with aspirin, 11 +- 5.4 IR neurons per slide in rats treated with acetaminophen, and 4 +- 3.7 IR neurons per slide in saline-treated control rats, P < 0.001]. Sodium Chloride 305-311 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-76 26515740-4 2017 Reductions in c-Fos-related immunoreactivity were found in dorsomedial striatum (DMS) but not dorsolateral striatum after exposure to the METH context suggesting this effect reflected a loss specifically in goal-directed control in the METH context. Methamphetamine 138-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 26515740-4 2017 Reductions in c-Fos-related immunoreactivity were found in dorsomedial striatum (DMS) but not dorsolateral striatum after exposure to the METH context suggesting this effect reflected a loss specifically in goal-directed control in the METH context. Methamphetamine 236-240 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 26515740-5 2017 This reduction in c-Fos was localized to non-enkephalin-expressing neurons in the DMS, likely dopamine D1-expressing direct pathway neurons, suggesting a relative change in control by the D1-direct versus D2-indirect pathways originating in the DMS may have been induced by METH-context exposure. Dopamine 94-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 26515740-5 2017 This reduction in c-Fos was localized to non-enkephalin-expressing neurons in the DMS, likely dopamine D1-expressing direct pathway neurons, suggesting a relative change in control by the D1-direct versus D2-indirect pathways originating in the DMS may have been induced by METH-context exposure. Methamphetamine 274-278 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 28225852-4 2017 The aim of this study was to verify the effects of a tryptophan-enriched diet on immunoreactivity to Fos-protein in the rat brain. Tryptophan 53-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-104 28225852-7 2017 Treatment with a tryptophan-enriched diet increased Fos-ir in the prefrontal cortex, nucleus accumbens, paraventricular hypothalamus, arcuate and ventromedial hypothalamus, dorsolateral and dorsomedial periaqueductal grey and dorsal and median raphe nucleus. Tryptophan 17-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 27377794-4 2017 We therefore assessed the effect of intrastriatal retigabine administration on striatal neuronal excitability in the rat determined by c-Fos immunoreactivity, a marker of neuronal activation. ezogabine 50-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 27377794-5 2017 When retigabine was applied locally in the striatum, this resulted in a marked reduction in the number of c-Fos-positive neurons after a strong excitatory striatal stimulus induced by acute systemic haloperidol administration in the rat. ezogabine 5-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 27377794-5 2017 When retigabine was applied locally in the striatum, this resulted in a marked reduction in the number of c-Fos-positive neurons after a strong excitatory striatal stimulus induced by acute systemic haloperidol administration in the rat. Haloperidol 199-210 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 27743985-6 2016 Naltrexone (20mg/kg) decreased tyrosine hydroxylase mRNA in the ventral tegmental area and Fos and Drd5 mRNA in NAc shell of HVR, but not LVR, rats. Naltrexone 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-94 27497640-8 2016 RESULTS: Microarray gene expression analysis showed that Pnoc, Cacfd1, Fos, Igll1, Lcn2, and Syk were among the most downregulated genes in the Qi Teng Xiao Zhuo granule group compared with the adriamycin treated group, whereas Cyp2c7, Hsd3b6, Acsm5, and Ugt2b15 were significantly upregulated. Doxorubicin 194-204 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 27491589-0 2016 Nicotinic receptor blockade decreases fos immunoreactivity within orexin/hypocretin-expressing neurons of nicotine-exposed rats. Nicotine 106-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 27847472-6 2016 Intracerebroventricular administration of astroglial toxin L-a-aminoadipate (L-AA) and c-Fos antisense oligodeoxy nucleotides (ASO) both decreased RWIS-induced gastric mucosal damage. Oligodeoxyribonucleotides 103-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 27847472-6 2016 Intracerebroventricular administration of astroglial toxin L-a-aminoadipate (L-AA) and c-Fos antisense oligodeoxy nucleotides (ASO) both decreased RWIS-induced gastric mucosal damage. 1,5-anhydroglucitol 127-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 27915481-10 2016 Tamsulosin suppressed contraction pressure and time while inhibiting c-Fos and NGF expressions. Tamsulosin 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 27436722-6 2016 Systemic buspirone (3 mg/kg) induced anxiogenic effects in elevated plus maze, light-dark box and open field exploration paradigms in rats and strongly activated the NI, as reflected by c-Fos expression. Buspirone 9-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 186-191 27260496-9 2016 OVX increased the expression of c-Fos (a neuronal marker of pain) in the thalamus; calcitonin strongly reversed this effect, and alendronate moderately reversed this effect. Alendronate 129-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 27566488-10 2016 Finally, we used c-Fos immunohistochemistry to identify the central mechanisms of the acute and repeated MK212 effects on maternal behavior. 6-chloro-2-(1-piperazinyl)pyrazine 105-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 27566488-11 2016 Acute MK212 (2.0mg/kg) disrupted pup retrieval and concurrently decreased c-Fos expression in the ventral part of lateral septal nucleus (LSv), MPOA, dentate gyrus (DG) and dorsal raphe (DR), but increased it in the central amygdala (CeA). 6-chloro-2-(1-piperazinyl)pyrazine 6-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 27566488-12 2016 Five days of repeated MK212 (2.0mg/kg) treatment produced a persistent disruption of pup retrieval and only decreased c-Fos expression in the DR. 6-chloro-2-(1-piperazinyl)pyrazine 22-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 27364433-10 2016 The marking of c-Fos in response to fenfluramine in the hypothalamic and raphe nuclei revealed, an especially lower activation of the PVN, MnR and DR compared intra-group. Fenfluramine 36-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 27826222-14 2016 The investigation of Fos expression in the brain showed neuronal activation in several brain nuclei such as the supraoptic nucleus, arcuate nucleus, locus coeruleus and nucleus of the solitary tract of ABA compared to AL rats. alisol B 23-acetate 202-205 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 27689413-7 2016 Immunohistochemistry showed that Cap evoked a significantly greater number of cells that were immunoreactive for c-Fos, a marker of nociceptive activation, in the trigeminal subnucleus caudalis/upper cervical cord in the HE group than in the LE group. Helium 221-223 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 27689413-7 2016 Immunohistochemistry showed that Cap evoked a significantly greater number of cells that were immunoreactive for c-Fos, a marker of nociceptive activation, in the trigeminal subnucleus caudalis/upper cervical cord in the HE group than in the LE group. Leu-Glu 242-244 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 27343381-11 2016 Rotigotine induced Fos expression in the caudate-putamen and SCH39166 completely blocked it. rotigotine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 27507424-0 2016 Rats showing low and high sensitization of frequency-modulated 50-kHz vocalization response to amphetamine differ in amphetamine-induced brain Fos expression. Amphetamine 95-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-146 27636578-14 2016 Alendronate treatment inhibited fracture-induced increases in hindpaw inflammatory mediators, reducing postfracture levels of tumor necrosis factor by 43% +- 9%, IL-1 by 60% +- 9%, IL-6 by 56% +- 14%, and NGF by 37% +- 14%, but had no effect on increased spinal cord Fos expression, or skin and sciatic nerve immunocomplex deposition. Alendronate 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 267-270 27507424-0 2016 Rats showing low and high sensitization of frequency-modulated 50-kHz vocalization response to amphetamine differ in amphetamine-induced brain Fos expression. Amphetamine 117-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-146 27507424-4 2016 We compared amphetamine-induced Fos expression in 16 brain regions considered important for the development of addiction between rats preselected for low and high sensitization of the response and next given nine daily amphetamine doses followed by a 2-week withdrawal and final amphetamine challenge. Amphetamine 12-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 27507424-6 2016 Compared to those in amphetamine-untreated controls, Fos-positive nuclei counts were significantly and markedly (2-6 times) higher in 12 regions in high-sensitized rats, whereas in low-sensitized rats they were significantly higher in the cingulate cortex and dorsomedial striatum only. Amphetamine 21-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 27656032-11 2016 Expression of hm4D-mCherry was specific to VTA dopamine cells and CNO blocked excitation and mating-induced cFos expression in VTA dopamine cells. Dopamine 131-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-112 27543844-6 2016 Alcohol decreased c-Fos IR in the mPFC, IC, Rh and AcbC. Alcohols 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 27060631-13 2016 The compound of paeoniflorin, a major bioactive compound in SFZYD, was proved to regulate the MAPK signaling pathway by inhibiting the expression of IL-1beta, IL-2, IL-10, IL-12, TNFalpha, INFgamma, c-jun and c-fos in PHA stimulated-PBMC. peoniflorin 16-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 209-214 27429160-4 2016 ARC neuronal activity, as determined by c-Fos immunoreactivity, was increased after a hypoglycemic stimulus by 2-deoxyglucose (2DG). Deoxyglucose 127-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 27619785-6 2016 Regarding the effect of Milnacipran administration, the number of c-Fos-positive cells was significantly decreased at all region of the L6 spinal cord in normal rats (P < 0.05), while this reduction was not observed in SCI rats. Milnacipran 24-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 27373591-7 2016 In contrast to previous literature with rats, swim stress brought a significant increase in c-Fos expression in the lateral septal nucleus and some other regions in the hypothalamus (the intermediate hypothalamic area, the paraventricular and arcuate nucleus) only in the imipramine-pretreated group, which has not been observed previously. Imipramine 272-282 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 27238894-6 2016 Less fos activation in the amygdala was observed in both enriched groups following a water escape task whereas an increase in fos activation in the nucleus accumbens was observed in the natural-enriched animals. Water 85-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 5-8 27583636-0 2016 Simultaneous Detection of c-Fos Activation from Mesolimbic and Mesocortical Dopamine Reward Sites Following Naive Sugar and Fat Ingestion in Rats. Dopamine 76-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 27583636-0 2016 Simultaneous Detection of c-Fos Activation from Mesolimbic and Mesocortical Dopamine Reward Sites Following Naive Sugar and Fat Ingestion in Rats. Sugars 114-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 27583636-1 2016 This study uses cellular c-fos activation to assess effects of novel ingestion of fat and sugar on brain dopamine (DA) pathways in rats. Dopamine 115-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 27583636-5 2016 Thus, sugar intake elicits DA release and increases c-fos-like immunoreactivity (FLI) from individual VTA DA projection zones including the nucleus accumbens (NAC), amygdala (AMY) and medial prefrontal cortex (mPFC) as well as the dorsal striatum. Sugars 6-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 27535909-8 2016 In adult Fos-lacZ transgenic rats, selective inactivation of nicotine-cue-activated Fos neurons in central amygdala, but not orbitofrontal cortex, decreased "incubated" nicotine seeking on withdrawal day 14. Nicotine 61-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 27535909-8 2016 In adult Fos-lacZ transgenic rats, selective inactivation of nicotine-cue-activated Fos neurons in central amygdala, but not orbitofrontal cortex, decreased "incubated" nicotine seeking on withdrawal day 14. Nicotine 61-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 27535909-8 2016 In adult Fos-lacZ transgenic rats, selective inactivation of nicotine-cue-activated Fos neurons in central amygdala, but not orbitofrontal cortex, decreased "incubated" nicotine seeking on withdrawal day 14. Nicotine 169-177 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 27535909-13 2016 Here, we used a rat model of incubation of drug craving, the neuronal activity marker Fos, and the Daun02 chemogenetic inactivation method to demonstrate that incubation of nicotine craving is also observed after adolescent-onset nicotine self-administration and that neuronal ensembles in the central nucleus of the amygdala play a critical role in this incubation in adult rats. Nicotine 173-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 27535915-2 2016 These regions express Fos (a marker of neural activity) during cue-induced reinstatement of cocaine seeking, but only subpopulations of neurons within these regions drive drug seeking. Cocaine 92-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-25 27535915-4 2016 In rats, we examined Fos expression during cue-induced reinstatement of cocaine- and sucrose-seeking in prelimbic cortex (PL), infralimbic cortex (IL), BLA, and vSub neurons that project to NAc core (NAcC) or NAc shell (NAcSh). Cocaine 72-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 27535915-4 2016 In rats, we examined Fos expression during cue-induced reinstatement of cocaine- and sucrose-seeking in prelimbic cortex (PL), infralimbic cortex (IL), BLA, and vSub neurons that project to NAc core (NAcC) or NAc shell (NAcSh). Sucrose 85-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 27535915-5 2016 Neurons in PL, BLA, and vSub that project to NAcC, but not NAcSh, expressed Fos during cue-induced cocaine seeking, but not sucrose seeking. Cocaine 99-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 27535915-12 2016 Prelimbic cortex (PL) projections to nucleus accumbens core (NAcC) uniquely expressed Fos in a manner that positively correlated with cocaine-seeking, but not sucrose-seeking, behavior. Cocaine 134-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 27388762-10 2016 Both EtOH- and LiCl-induced CTA significantly enhanced cFos expression in the RMTg and LHb but not the hippocampus. Ethanol 5-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-59 27388762-10 2016 Both EtOH- and LiCl-induced CTA significantly enhanced cFos expression in the RMTg and LHb but not the hippocampus. Lithium Chloride 15-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-59 27388762-10 2016 Both EtOH- and LiCl-induced CTA significantly enhanced cFos expression in the RMTg and LHb but not the hippocampus. Chlorotrianisene 28-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-59 27388762-13 2016 In addition, cFos expression in the RMTg was positively correlated with LHb cFos. 5-[[[(2~{r},3~{s},4~{r},5~{r})-5-(6-Aminopurin-9-Yl)-3,4-Bis(Oxidanyl)oxolan-2-Yl]methylamino]methyl]-4-Azanyl-1-(Methoxymethyl)pyrimidin-2-One 72-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-17 27388762-13 2016 In addition, cFos expression in the RMTg was positively correlated with LHb cFos. 5-[[[(2~{r},3~{s},4~{r},5~{r})-5-(6-Aminopurin-9-Yl)-3,4-Bis(Oxidanyl)oxolan-2-Yl]methylamino]methyl]-4-Azanyl-1-(Methoxymethyl)pyrimidin-2-One 72-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-80 27388762-15 2016 Furthermore, increased cFos expression in the RMTg following EtOH-induced CTA suggests that this region plays a role in signaling alcohol"s aversive properties. Ethanol 61-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-27 27388762-15 2016 Furthermore, increased cFos expression in the RMTg following EtOH-induced CTA suggests that this region plays a role in signaling alcohol"s aversive properties. Alcohols 130-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-27 27456222-6 2016 Immunohistochemistry study indicated that application of CaCl2 (25 or 50 nmol) in the DRN significantly increased c-Fos expression ratio in wake-promoting neurons including serotonergic neurons in the DRN, noradrenergic neurons in the locus coeruleus, and orxinergic neurons in the perifornical nucleus, but decreased c-Fos expression ratio of GABAergic sleep-promoting neurons in the ventrolateral preoptic nucleus. Calcium Chloride 57-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 27456222-6 2016 Immunohistochemistry study indicated that application of CaCl2 (25 or 50 nmol) in the DRN significantly increased c-Fos expression ratio in wake-promoting neurons including serotonergic neurons in the DRN, noradrenergic neurons in the locus coeruleus, and orxinergic neurons in the perifornical nucleus, but decreased c-Fos expression ratio of GABAergic sleep-promoting neurons in the ventrolateral preoptic nucleus. Calcium Chloride 57-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 318-323 27102341-5 2016 In contrast to this, foot-shocked rats (emitting the 22-kHz vocalizations) had the c-Fos positivity increased markedly in the PAG and only slightly in the AC. pag 126-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 27102341-6 2016 The expression of c-Fos also increased in the IC, AC, and in the PAG in animals exposed to the artificial call-like stimuli, when compared to controls; however, the increase was much less pronounced. pag 65-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 27471467-5 2016 Brain mapping analysis of Fos expression, a biological marker of neural excitation, revealed that the seizure threshold level of PTZ region-specifically elevated Fos expression in the amygdala in Sv2a(L174Q) rats. Pentylenetetrazole 129-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 27471467-5 2016 Brain mapping analysis of Fos expression, a biological marker of neural excitation, revealed that the seizure threshold level of PTZ region-specifically elevated Fos expression in the amygdala in Sv2a(L174Q) rats. Pentylenetetrazole 129-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-165 26743854-7 2016 Visceral hypersensitivity was associated with an increase in the expression of c-fos, corticotropin-releasing factor (CRF) protein and mRNA in PVN, which could be prevented by intra-PVN infusion of lidocaine or small interfering RNA targeting the CRF gene. Lidocaine 198-207 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 27367964-5 2016 When the prodrug Daun02 is injected into the brains of these rats 90 min after a behavior (e.g., drug-seeking) or cue exposure, then Daun02 is converted into daunorubicin by beta-gal, which selectively inactivates Fos- and beta-gal-expressing neurons that were activated 90 min before the Daun02 injection. Daun02 17-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 214-217 27367964-5 2016 When the prodrug Daun02 is injected into the brains of these rats 90 min after a behavior (e.g., drug-seeking) or cue exposure, then Daun02 is converted into daunorubicin by beta-gal, which selectively inactivates Fos- and beta-gal-expressing neurons that were activated 90 min before the Daun02 injection. Daunorubicin 158-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 214-217 26873551-6 2016 Application of DEX leads to increased activity of CA1/CA3 hippocampal MAP2-positive neurons, as determined by c-Fos expression at 0.5-1h after DEX injection. Dexamethasone 15-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 26979294-8 2016 As expected, compared with Controls, Paired rats administered IP amphetamine subsequently showed a conditioned locomotor response when challenged with saline in the open field, an effect accompanied by an increase in c-Fos+ neurons in the medial NAcc. Amphetamine 65-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 217-222 26979294-11 2016 Together, these results suggest a role for c-Fos+ neurons in the medial NAcc and rapid changes in the morphology of their dendritic spines in the expression of conditioning evoked by amphetamine-paired contextual stimuli. Amphetamine 183-194 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 27203571-7 2016 administration of CCK-8 (50 mug/kg) resulted in marked increases in the number of nesfatin-1-IR neurones expressing Fos-immunoreactivity in the SON, PVN, AP and NTS but not in the ARC or LHA. cholecystokinin 8 18-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 27091613-5 2016 In the dorsomedial part of the bed nucleus of the stria terminalis (dmBNST), SI-induced c-Fos expression was observed only in PCP-treated rats. pcp 126-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 27091613-6 2016 Interestingly, URB597 in PCP-treated rats restored a similar c-Fos expression pattern as observed in saline-treated rats: activation of the orbitofrontal cortex, inhibition of the central amygdala and suppression of activation of the dmBNST. cyclohexyl carbamic acid 3'-carbamoylbiphenyl-3-yl ester 15-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 27091613-6 2016 Interestingly, URB597 in PCP-treated rats restored a similar c-Fos expression pattern as observed in saline-treated rats: activation of the orbitofrontal cortex, inhibition of the central amygdala and suppression of activation of the dmBNST. pcp 25-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 27288111-4 2016 Similar to an IS infusion, IS-RIZ treatment induced nociception-related behaviours in Sprague-Dawley rats and increased Fos expression in the cortex and trigeminal pathway, whereas the RIZ injection alone did not. rizatriptan 30-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-123 26919917-2 2016 In this study, we observed c-Fos expressions in orexinergic neurons following isoflurane inhalation (for 0, 30, 60, and 120min) and at the time when the righting reflex returned after the cessation of anesthesia. Isoflurane 78-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 26919917-6 2016 The results showed that the numbers of c-Fos-immunoreactive orexinergic neurons in the hypothalamus decreased over time with continued isoflurane inhalation, but restored at emergence. Isoflurane 135-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 27060631-15 2016 And the mechanisms were closely related with the regulation of the MAPK pathway by reduction in phosphorylated forms of the three MAPK (ERK1/2, p38 and JNK) and down regulation of c-jun and c-fos by paeoniflorin. peoniflorin 199-211 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 190-195 27094734-0 2016 Prenatal valproic acid exposure disrupts tonotopic c-Fos expression in the rat brainstem. Valproic Acid 9-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 27556131-0 2016 Corrigendum to "Prenatal valproic acid exposure disrupts tonotopic c-Fos expression in the rat brainstem" [Neuroscience 311 (2015) 349-361]. Valproic Acid 25-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 27094734-6 2016 Herein, we have subjected control and VPA-exposed animals to 4- or 16-kHz tones and examined neuronal activation with immunohistochemistry for c-Fos. Valproic Acid 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-148 27114001-9 2016 We also used c-Fos immunohistochemistry as a marker of neuronal activation and found that AcbC OT injection increases activation of the AcbC itself, as well as of two feeding-related sites: the hypothalamic paraventricular and supraoptic nuclei. 1-aminocyclobutanecarboxylic acid 90-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 27030665-5 2016 Moreover, the number of Fos-expressing neurons was significantly and positively correlated in the two responsive regions with terminal blood glucose concentrations. Glucose 141-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 27030665-7 2016 We found the expected and highly significant increase in Fos in the neuroendocrine PVH, which was negatively correlated to terminal blood glucose concentrations, but no significant differences were seen in any part of the PAG. Glucose 138-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 27030665-8 2016 Our results show that there are distinct populations of VMH neurons whose Fos expression is suppressed by hypoglycemia, and their numbers correlate with blood glucose. Glucose 159-166 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 26068175-0 2016 Effects of acute or chronic environmental enrichment on regional Fos protein expression following sucrose cue-reactivity testing in rats. Sucrose 98-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 25976757-5 2016 Pretreatment with metyrapone decreased freezing and Fos expression in these areas. Metyrapone 18-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 26703089-10 2016 Differential expression profiles of the immediate early gene cFos indicated hippocampal involvement in the inference process while localized microinfusions of the muscarinic antagonist, scopolamine, into the dorsal hippocampus caused rats to behave as if only one light was present. Scopolamine 186-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-65 27144381-4 2016 (ii) OXT-mRFP1 neurones in the anterior parvocellular PVN, which may project to the dorsal horn of the spinal cord, were activated by s.c. injection of formalin, as indicated by a significant increases of Fos-IR and mRFP1 intensity intensity. Formaldehyde 152-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 205-208 27103167-14 2016 Inactivation of CREB and c-fos by MC1568 likely contributes to this detrimental effect. MC1568 34-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 27020784-5 2016 In experiment 2, we determined potential central sites of action by analyzing effects of prazosin (1 mg/kg) on yohimbine (1.25 mg/kg)-induced Fos expression. Prazosin 89-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-145 27020784-5 2016 In experiment 2, we determined potential central sites of action by analyzing effects of prazosin (1 mg/kg) on yohimbine (1.25 mg/kg)-induced Fos expression. Yohimbine 111-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-145 27020784-8 2016 Systemic prazosin reduced yohimbine-induced Fos expression in the prefrontal cortex, accumbens shell, ventral bed nucleus of the stria terminalis, and basolateral amygdala (46-67 % decreases). Prazosin 9-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 27020784-8 2016 Systemic prazosin reduced yohimbine-induced Fos expression in the prefrontal cortex, accumbens shell, ventral bed nucleus of the stria terminalis, and basolateral amygdala (46-67 % decreases). Yohimbine 26-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 27234303-7 2016 RESULTS: c-Fos protein expression and the frequency of both spontaneous action potential firings and spontaneous excitatory postsynaptic currents were higher in LHb neurons of ethanol-withdrawn rats compared to their ethanol-naive counterparts. 5-[[[(2~{r},3~{s},4~{r},5~{r})-5-(6-Aminopurin-9-Yl)-3,4-Bis(Oxidanyl)oxolan-2-Yl]methylamino]methyl]-4-Azanyl-1-(Methoxymethyl)pyrimidin-2-One 161-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 27234303-7 2016 RESULTS: c-Fos protein expression and the frequency of both spontaneous action potential firings and spontaneous excitatory postsynaptic currents were higher in LHb neurons of ethanol-withdrawn rats compared to their ethanol-naive counterparts. Ethanol 176-183 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 27234303-7 2016 RESULTS: c-Fos protein expression and the frequency of both spontaneous action potential firings and spontaneous excitatory postsynaptic currents were higher in LHb neurons of ethanol-withdrawn rats compared to their ethanol-naive counterparts. Ethanol 217-224 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 26821289-9 2016 In addition, an increase in the percentage of CRF-containing neurons co-localized with c-Fos was observed in the dmBNST after EPM exposure. dmbnst 113-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 27118833-6 2016 Noteworthy, the H1 receptor antagonist pyrilamine disrupted IA memory retrieval in rats, thus strongly supporting an active involvement of endogenous histamine; 90 min after the retention test, c-Fos-positive neurons were significantly fewer in the CA1s of a-FMHis-treated rats that displayed amnesia compared with in the control group. Pyrilamine 39-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 194-199 27275127-7 2016 Intrathecal injection of SCH772984 has analgesic effects on rats with bone cancer pain, and the effects enhance with increasing dose; intrathecal injection of SCH772984 10 mug could greatly reduce the expression of spinal dorsal horn Fos protein. SCH772984 25-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 234-237 27275127-7 2016 Intrathecal injection of SCH772984 has analgesic effects on rats with bone cancer pain, and the effects enhance with increasing dose; intrathecal injection of SCH772984 10 mug could greatly reduce the expression of spinal dorsal horn Fos protein. SCH772984 159-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 234-237 26438555-8 2016 There was a significant increase in the mean number of c-Fos neurons in the PAG, RMg, and TNC in Model versus Control groups (p<0.001); however, this was significantly attenuated by EA treatment (p<0.001). pag 76-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 26850920-3 2016 In the first experiment, animals were maintained on caffeinated drinking water or normal tap water for 14 days and were then tested for behavioral and striatal c-Fos response to amphetamine (1.5 mg/kg). Amphetamine 178-189 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 26861675-9 2016 The ERK-CREB-Fos pathway and the NMDA receptor NR2B subunits in the NAc were involved in the cocaine-induced behavioral sensitization. Cocaine 93-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 27239846-0 2016 [Influence of dexamethasone on the expression of immediate early genes c-fos and c-jun in different regions of the neonatal brain]. Dexamethasone 14-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 26874560-14 2016 Adolescent caffeine consumption increased basal c-fos mRNA in the paraventricular nucleus of the hypothalamus. Caffeine 11-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 26876759-0 2016 Contrasting regional Fos expression in adolescent and young adult rats following acute administration of the antidepressant paroxetine. Paroxetine 124-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 26876759-4 2016 Paroxetine induced widespread Fos expression in both adolescent and young adult rats. Paroxetine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 26876759-10 2016 These results indicate a different c-Fos signature of acute paroxetine in adolescent rats, with greater activation in key mesolimbic and serotonergic regions, but a more subdued cortical, brainstem and hypothalamic response. Paroxetine 60-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 26274988-8 2016 RESULTS: MCS was found to modulate c-fos and serotonin expression. mcs 9-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 27239846-5 2016 The dexamethasone-induced shift of the ratio of c-jun to c-fos transcript levels in the brainstem of neonatal rats towards a predominance of c-jun reported for the first time in the present work may induce the expression of genes that contain AP-1 response elements in the promoters, since the glucocorticoid receptor can be involved in protein-protein interactions with the Jun/Jun homodimer of the AP-1 complex. Dexamethasone 4-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 26289145-7 2016 Following SD, Fos co-expression in Hcrt, HA, and ACh neurons (but not in 5HT neurons) was consistently elevated in VEH- and ALM-treated rats, whereas Fos expression in these neuronal groups was unaffected by SD in ZOL-treated rats. Zolpidem 214-217 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 26541329-9 2016 Finally, we demonstrated that inhibition of MAGL reduced VIC Fos immunoreactivity in response to LiCl treatment. Lithium Chloride 97-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 26645585-4 2016 Furthermore, DPAA overpoweringly increased the phosphorylation of three major mitogen-activated protein kinases (MAPKs) (ERK1/2, p38MAPK, and SAPK/JNK), which indicated MAPK activation, and subsequently induced expression and/or phosphorylation of transcription factors (Nrf2, CREB, c-Jun, and c-Fos) in cultured rat cerebellar astrocytes. diphenylarsinic acid 13-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 294-299 26655477-8 2016 These results suggest that multiple phenotypic regions are mediated by NMDAR and Fos/DeltaFosB during morphine withdrawal, such as the motivational (AcbC, AcbSh), limbic (CeC, VTA) and executive (Cg) system pathways, and may be the primary targets of NMDAR and Fos/DeltafosB that impact morphine withdrawal behaviors. Morphine 102-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-84 26655477-8 2016 These results suggest that multiple phenotypic regions are mediated by NMDAR and Fos/DeltaFosB during morphine withdrawal, such as the motivational (AcbC, AcbSh), limbic (CeC, VTA) and executive (Cg) system pathways, and may be the primary targets of NMDAR and Fos/DeltafosB that impact morphine withdrawal behaviors. Morphine 102-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 26655477-8 2016 These results suggest that multiple phenotypic regions are mediated by NMDAR and Fos/DeltaFosB during morphine withdrawal, such as the motivational (AcbC, AcbSh), limbic (CeC, VTA) and executive (Cg) system pathways, and may be the primary targets of NMDAR and Fos/DeltafosB that impact morphine withdrawal behaviors. Morphine 287-295 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-84 27239846-2 2016 The aim of the present work was to assess the levels of mRNA encoded by genes c-jun and c-fos and the ratio of expression levels of these genes in various regions of the neonatal rat brain after the administration of dexamethasone, a selective ligand of the glucocorticoid receptor. Dexamethasone 217-230 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 27239846-3 2016 The level of mRNA encoded by the immediate early gene c-fos in the hippocampus and prefrontal cortex of 3-day-old rat pups was elevated at 30, 60, and 120 min after dexamethasone administration. Dexamethasone 165-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 26786746-0 2016 Alcohol consumption increases locomotion in an open field and induces Fos-immunoreactivity in reward and approach/withdrawal-related neurocircuitries. Alcohols 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 26845170-7 2016 In the present work, male rats rendered motivated to obtain water, sex, or amphetamine showed an increase in Fos-ir of histaminergic neurons in appetitive behaviors directed to get those reinforcers. Water 60-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-112 26845170-7 2016 In the present work, male rats rendered motivated to obtain water, sex, or amphetamine showed an increase in Fos-ir of histaminergic neurons in appetitive behaviors directed to get those reinforcers. Amphetamine 75-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-112 26845170-8 2016 However, during appetitive tests to obtain sex, or drug in amphetamine-conditioned rats, Fos expression increased in most other ascending arousal system nuclei, including the orexin neurons in the lateral hypothalamus, dorsal raphe, locus coeruleus and laterodorsal tegmental neurons, but not in the ventral tegmental area, which showed no Fos-ir increase in any of the 3 conditions. Amphetamine 59-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 26845170-8 2016 However, during appetitive tests to obtain sex, or drug in amphetamine-conditioned rats, Fos expression increased in most other ascending arousal system nuclei, including the orexin neurons in the lateral hypothalamus, dorsal raphe, locus coeruleus and laterodorsal tegmental neurons, but not in the ventral tegmental area, which showed no Fos-ir increase in any of the 3 conditions. Amphetamine 59-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 340-343 26775553-9 2016 Acute ethanol treatment significantly increased Fos immunoreactivity in the BNST and the central amygdala. Ethanol 6-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 27065810-7 2016 Water deprivation significantly reduced freezing and immobility behaviors evoked by innate fear and behavioral despair, respectively, accompanied by decreased Fos expression in the lateral habenula. Water 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-162 26786746-9 2016 Additionally, alcohol intake increased Fos-immunoreactivity (Fos-ir) in the prefrontal cortex, in the shell region of the nucleus accumbens, in the medial and central amygdala, in the bed nucleus of the stria terminalis, in the septal region, and in the paraventricular and dorsomedial hypothalamus, structures that have been linked to reward and to approach/withdrawal behavior. Alcohols 14-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 26786746-9 2016 Additionally, alcohol intake increased Fos-immunoreactivity (Fos-ir) in the prefrontal cortex, in the shell region of the nucleus accumbens, in the medial and central amygdala, in the bed nucleus of the stria terminalis, in the septal region, and in the paraventricular and dorsomedial hypothalamus, structures that have been linked to reward and to approach/withdrawal behavior. Alcohols 14-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 26391065-10 2016 MK 801 prevented mating-induced cFos expression in NAc shell and core. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-36 26216055-5 2016 The number of formalin-induced FRB 15-30 min after the formalin injection was significantly higher in IB4-Sap-treated rats than in Sal- or Bl-Sap-treated rats, and was associated with an increase in c-Fos-IR cells. Formaldehyde 14-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-204 26601772-7 2016 Hypercapnia induced by 7% CO2 increased the number of c-Fos/TH-immunoreactive (ir) neurons in the LC of all groups when compared to air exposure. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 26-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 26216055-5 2016 The number of formalin-induced FRB 15-30 min after the formalin injection was significantly higher in IB4-Sap-treated rats than in Sal- or Bl-Sap-treated rats, and was associated with an increase in c-Fos-IR cells. ib4-sap 102-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-204 26216055-6 2016 The systemic preadministration of the GABAA antagonist, bicuculline, and agonist, muscimol, had stronger decreasing effects on FRB and c-Fos-IR cells in IB4-Sap-treated rats than the preadministration of Sal, whereas the opposite effects were observed in Sal- and Bl-Sap-treated rats. Bicuculline 56-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 26216055-6 2016 The systemic preadministration of the GABAA antagonist, bicuculline, and agonist, muscimol, had stronger decreasing effects on FRB and c-Fos-IR cells in IB4-Sap-treated rats than the preadministration of Sal, whereas the opposite effects were observed in Sal- and Bl-Sap-treated rats. Muscimol 82-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 27069541-9 2016 Compared with the model group, rats given Dex had (1) shorter escape latencies, (2) more platform crossings, (3) fewer relaxin-3 and c-fos positive neurons in the hippocampal CA1 area, (4) upregulation of Bcl-2, (5) downregulation of Fas, caspase-8, and caspase-9 proteins, and (6) decreased neuroapoptosis in the hippocampus. Dexmedetomidine 42-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 27543774-13 2016 In immunohistochemical sections, c-FOS positivity was decreased in the NTG and especially the TG. Nitroglycerin 71-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 27430911-5 2016 In a kainic acid rat model, intraperitoneally administering baicalein to rats before the kainic acid intraperitoneal injection substantially attenuated kainic acid-induced neuronal cell death, c-Fos expression, and the activation of the mammalian target of rapamycin in the hippocampus. baicalein 60-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-198 26727528-0 2016 Alcohol Induces Parallel Changes in Hippocampal Histone H3 Phosphorylation and c-Fos Protein Expression in Male Rats. Alcohols 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 26727528-10 2016 Subsequent examination of c-fos, a gene known to be regulated by H3S10ph, revealed that EtOH and withdrawal-associated changes in c-fos closely paralleled changes in H3S10ph. Ethanol 88-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 26727528-10 2016 Subsequent examination of c-fos, a gene known to be regulated by H3S10ph, revealed that EtOH and withdrawal-associated changes in c-fos closely paralleled changes in H3S10ph. Ethanol 88-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 26727528-11 2016 CONCLUSIONS: These results suggest that H3S10ph regulates EtOH-mediated changes in c-fos expression, effects that likely have important implications for EtOH-induced changes in hippocampal neuronal plasticity. h3s10ph 40-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 26727528-11 2016 CONCLUSIONS: These results suggest that H3S10ph regulates EtOH-mediated changes in c-fos expression, effects that likely have important implications for EtOH-induced changes in hippocampal neuronal plasticity. Ethanol 58-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 26727528-11 2016 CONCLUSIONS: These results suggest that H3S10ph regulates EtOH-mediated changes in c-fos expression, effects that likely have important implications for EtOH-induced changes in hippocampal neuronal plasticity. Ethanol 153-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 26406609-7 2016 The hypertrophic phenotype (cardiomyocyte size, sarcomeric organization, total protein synthesis, c-fos expression) mediated by phenylephrine (10 muM), but not that by angiotensinII (1 muM) or IGF1 (20 ng mL(-1) ), was counteracted by the selective A1 receptor agonist, N6-cyclopentyladenosine. Phenylephrine 128-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 27543774-13 2016 In immunohistochemical sections, c-FOS positivity was decreased in the NTG and especially the TG. Thioguanine 72-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 26522737-4 2016 Additionally, the present data provide evidence that the central GLP-1-producing preproglucagon neurons in the nucleus tractus solitarius (NTS) of the caudal brainstem are activated by cisplatin during the delayed phase of chemotherapy-induced nausea, as cisplatin led to a significant increase in c-Fos immunoreactivity in NTS GLP-1-immunoreactive neurons. Cisplatin 185-194 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 298-303 26883904-0 2016 Stress alters asenapine-induced Fos expression in the Meynert"s nucleus: response of adjacent hypocretin and melanin-concentrating hormone neurons in rat. asenapine 14-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 26883904-1 2016 OBJECTIVE: Asenapine (ASE), an atypical antipsychotic drug used in the treatment of schizophrenia, induces Fos expression in forebrain. asenapine 11-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 26883904-1 2016 OBJECTIVE: Asenapine (ASE), an atypical antipsychotic drug used in the treatment of schizophrenia, induces Fos expression in forebrain. asenapine 22-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 26536818-6 2015 Complementary neural analysis revealed that decreased cue-induced feeding under SB-334867 increased Fos expression in mPFC and paraventricular thalamus. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 80-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-103 26478151-0 2016 Ginkgo biloba Extract (EGb 761 ) Inhibits Glutamate-induced Up-regulation of Tissue Plasminogen Activator Through Inhibition of c-Fos Translocation in Rat Primary Cortical Neurons. Glutamic Acid 42-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-133 26433325-5 2016 We also found that AMPH administration completely blocked the forced swim-induced expression of the corticotropin-releasing hormone (hnCRH) and it partially reduced c-fos expression in the paraventricular nucleus of the hypothalamus (PVN). Amphetamine 19-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 26733817-10 2015 Finally, intra-IC infusion of U0126 obviously decreased Fos expression in the Vc, accompanied by the alleviation of both nociceptive behavior and negative emotions. U 0126 30-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 26454025-6 2015 MK-801-induced dopamine release increase in acetylcholinesterase (AChE), mono amine oxidase (MAO) activity and increase in c-fos expression were also investigated. Dizocilpine Maleate 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 26518464-0 2015 Prenatal valproic acid exposure disrupts tonotopic c-Fos expression in the rat brainstem. Valproic Acid 9-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 26518464-6 2015 Herein, we have subjected control and VPA-exposed animals to 4 or 16 kHz tones and examined neuronal activation with immunohistochemistry for c-Fos. Valproic Acid 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-147 26400184-4 2015 Low-dose intracarotid artery infusion of RLX (155 pmol ml(-1) h(-1); 1.5 h) had minor transient effects on AP and activated neurons [increased Fos-immunoreactivity (IR)] in the SFO and in spinally projecting (19 +- 2%) and arginine-vasopressin (AVP)-IR (21 +- 5%) cells in the paraventricular nucleus of the hypothalamus of NP, but not pregnant (P), rats. Relaxin 41-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-146 26524411-1 2015 Intraperitoneal injections (ip) of lithium chloride at large doses induce c-Fos expression in the brain regions implicated in conditioned taste aversion (CTA) learning, and also activate the hypothalamic-pituitary-adrenal (HPA) axis and increase the plasma corticosterone levels in rats. Lithium Chloride 35-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 26524411-2 2015 A pharmacologic treatment blunting the lithium-induced c-Fos expression in the brain regions, but not the HPA axis activation, induced CTA formation. Lithium 39-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 26524411-6 2015 ip lithium consistently induced the brain c-Fos expression, the HPA activation and CTA formation regardless of the circadian activation of the HPA axis. Lithium 3-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 26524411-7 2015 Intracerebroventricular (icv) injections of lithium at day time also increased the brain c-Fos expression, activated the HPA axis and induced CTA acquisition. Lithium 44-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 26524411-8 2015 However, icv lithium at night, when the HPA axis shows its circadian activation, did not induce CTA acquisition nor activate the HPA axis, although it increased the brain c-Fos expression. Lithium 13-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-176 26363150-7 2015 M1 expression of two immediate-early genes (c-Fos and ARC) was strongly enhanced by either L-DOPA or SKF81297. Levodopa 91-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 26234469-5 2015 The study showed that after 40% food restriction in maternal dietary intake, the dipsogenic responses for both water and salt intake were not altered; Fos expression in brain areas investigated involved in hydromineral homeostasis control was always higher in the offspring in response to isoproterenol. Isoproterenol 289-302 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-154 28911484-7 2015 The aspartame treated MTX animals showed a marked increase in the c-fos, Hsp 70, iNOS Caspase 8, and JNK 3 protein expression in the liver from control and MTX control animals indicating the enhancement of stress and apoptosis. Aspartame 4-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 28911484-7 2015 The aspartame treated MTX animals showed a marked increase in the c-fos, Hsp 70, iNOS Caspase 8, and JNK 3 protein expression in the liver from control and MTX control animals indicating the enhancement of stress and apoptosis. Methotrexate 22-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 26539813-7 2015 Honokiol treatment normalized hippocampal and thalamic c-Fos and hippocampal alveus substance P receptor expression relative to controls at P21. honokiol 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 26616874-9 2015 LPS induced Fos protein expression in several areas of the brains of the control rats; however, higher numbers of Fos-positive cells were observed in the brains of the rats that were fed a fructose diet. Fructose 189-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 26492981-1 2015 This study examined the influence of propofol anesthesia on the expression of activity-regulated molecules (BDNF and c-Fos) and synaptic plasticity markers (synaptophysin, GAP-43, drebrin) in the frontal cortex and thalamus of 7-day-old (P7) rats. Propofol 37-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 28911484-6 2015 The aspartame treated MTX animals showed a marked increase in the inducible nitric oxide (iNOS), neuronal nitric oxide (nNOS), c-fos, Heat shock protein (Hsp) 70 Tumour necrosis Factor (TNF)alpha, caspase 8, c-jun N terminal kinases (JNK) 3 and Nuclear factor kappa B (NFkB) gene expression in the liver from control and MTX control animals. Aspartame 4-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 28911484-6 2015 The aspartame treated MTX animals showed a marked increase in the inducible nitric oxide (iNOS), neuronal nitric oxide (nNOS), c-fos, Heat shock protein (Hsp) 70 Tumour necrosis Factor (TNF)alpha, caspase 8, c-jun N terminal kinases (JNK) 3 and Nuclear factor kappa B (NFkB) gene expression in the liver from control and MTX control animals. Methotrexate 22-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 26298729-11 2015 BBG attenuated the increase in c-Fos-positive cells in the caudal trigeminal nucleus (TNC) after robust stimulation, but not after mild stimulation. coomassie Brilliant Blue 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 26044638-3 2015 Our studies confirmed that administration of MP-10 significantly elevates expression of the immediate early genes (IEG) c-fos, egr-1, and arc in rat striatum. 2-((4-(1-methyl-4-pyridin-4-yl-1H-pyrazol-3-yl)phenoxy)methyl)quinoline 45-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 26256420-5 2015 The D2 antagonist, haloperidol (0.3, 1, or 3 mg/kg, PO) produced an identical, regional pattern of c-Fos induction favoring the dorsolateral striatum of the rat. Haloperidol 19-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 26318100-7 2015 Acute midazolam administration significantly attenuated the neophobia and conditioned fear responses, decreased c-Fos expression in the LA and CeA, and increased alpha-2 subunit density in the CeA only in the HR group. Midazolam 6-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 25560759-5 2015 Acute tianeptine treatment selectively altered Fos expression within subdivisions of the central nucleus of the amygdala (CEA) in a bidirectional manner that varied in relation to ongoing activation within the capsular subdivision and its prefrontal and intra-amygdalar inputs. tianeptine 6-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 26649942-0 2015 A pivotal role of FOS-mediated BECN1/Beclin 1 upregulation in dopamine D2 and D3 receptor agonist-induced autophagy activation. Dopamine 62-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 26297864-9 2015 Corticosterone decreased maternal care, plasma oxytocin levels, milk consumption by the pups, the activation of oxytocinergic neurons in hypothalamic nuclei, and c-Fos immunoreactivity in MPOA neurons. Corticosterone 0-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 26171591-5 2015 In cell-dehydrated rats with no access to fluids, methysergide compared to vehicle increased Fos immunoreactivity in the medial nucleus of the solitary tract, area postrema and non-oxytocinergic cells of the ventral portion of the hypothalamic paraventricular nucleus (PVN). Methysergide 50-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 26299338-8 2015 RESULTS: Repeated amphetamine exposure (a) prevented the increase in sodium intake and Fos-IR cells in caudate-putamen and accumbens nucleus induced by ANG II i.c.v. Amphetamine 18-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 26386156-3 2015 The expression of c-fos was transiently induced by treatment of cells with high potassium (high K(+)), which evoked depolarization, or forskolin, an adenylate cyclase activator. Potassium 80-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 26386156-3 2015 The expression of c-fos was transiently induced by treatment of cells with high potassium (high K(+)), which evoked depolarization, or forskolin, an adenylate cyclase activator. Colforsin 135-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 26386156-4 2015 c-fos expression was persistently and synergistically induced by simultaneous treatment with high K(+) and forskolin via cAMP-response element (CRE). Colforsin 107-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 26386156-4 2015 c-fos expression was persistently and synergistically induced by simultaneous treatment with high K(+) and forskolin via cAMP-response element (CRE). Cyclic AMP 121-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 26434709-3 2015 Oral pretreatment with BD1047 dose-dependently reduced zymosan-induced thermal and mechanical hyperalgesia as well as spinal neuronal activation including increased immunoreactivity of Fos, protein kinase C (PKC) and "PKC-dependent phosphorylation of the NMDA receptor subunit 1" (pNR1). N-(2-(3,4-Dichlorphenyl)ethyl)-N,N',N'-trimethyl-1,2-ethandiamin 23-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 185-188 25608877-6 2015 Furthermore, we investigated whether URB937 affects NTG-induced c-Fos expression in the brain. Nitroglycerin 52-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 25608877-9 2015 Mapping of brain nuclei activated by NTG indicates that URB937 significantly reduces c-Fos expression in the nucleus trigeminalis caudalis and the locus coeruleus. Nitroglycerin 37-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 26225927-3 2015 Immunohistochemical techniques were utilized to detect the expression of c-Fos and p-p38 in SDH in the control and amiloride (100mug) groups. sdh 92-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 26225927-3 2015 Immunohistochemical techniques were utilized to detect the expression of c-Fos and p-p38 in SDH in the control and amiloride (100mug) groups. Amiloride 115-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 26225927-5 2015 Additionally, immunohistochemical experiments showed that the expression of c-Fos and p-p38 dramatically decreased in the superficial laminae of the ipsilateral SDH in the 100-mug amiloride group (P<0.01), whereas, there was no statistical significance on the contralateral side, compared with the control group. sdh 161-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 26225927-5 2015 Additionally, immunohistochemical experiments showed that the expression of c-Fos and p-p38 dramatically decreased in the superficial laminae of the ipsilateral SDH in the 100-mug amiloride group (P<0.01), whereas, there was no statistical significance on the contralateral side, compared with the control group. Amiloride 180-189 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 26225927-7 2015 It most likely reduces spinal neurons and microglia activation via inhibiting c-Fos and p-p38 MAPK in the SDH of rats. sdh 106-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 26003276-4 2015 Intraperitoenal injection of lithium chloride (0.15M, 3ml/kg or 12ml/kg) at night induced CTA formation and the HPA axis activation and increased c-Fos expression in both the parabrachial nucleus (PBN) and the nucleus tractus of solitarius (NTS) in a dose dependent manner. Lithium Chloride 29-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-151 26003276-5 2015 However, intracerebroventricular lithium (0.6M, 5microl) at night failed to induce CTA or the HPA axis activation, although it increased c-Fos expression in the PBN and NTS. Lithium 33-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 26459117-8 2015 PF induced higher Fos expression in the nucleus accumbens shell and core and in the prelimbic and infralimbic cortex of BEP rats compared to No PF controls. pf 0-2 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 26190766-0 2015 Brain regions associated with inverse incentive learning: c-Fos immunohistochemistry after haloperidol sensitization on the bar test in rats. Haloperidol 91-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 26190766-6 2015 c-Fos levels in the nucleus accumbens core and ventral pallidum were lower in the Paired group than in the Unpaired and Saline groups even though all groups received haloperidol on the test day. Sodium Chloride 120-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 26190766-6 2015 c-Fos levels in the nucleus accumbens core and ventral pallidum were lower in the Paired group than in the Unpaired and Saline groups even though all groups received haloperidol on the test day. Haloperidol 166-177 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 26190766-7 2015 Compared to the Saline group both the Paired and Unpaired groups showed evidence of lower c-Fos levels in the dorsal striatum and nucleus accumbens shell, possibly a result of daily haloperidol injections. Sodium Chloride 16-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 26190766-7 2015 Compared to the Saline group both the Paired and Unpaired groups showed evidence of lower c-Fos levels in the dorsal striatum and nucleus accumbens shell, possibly a result of daily haloperidol injections. Haloperidol 182-193 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 26320022-8 2015 AS30 in the ADX group attenuated the enhanced Fos expression but not the number of Fos+OT double labeled neurons in the PaMP. antisauvagine 30 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 26322574-0 2015 Antenatal Taurine Improves Intrauterine Growth-Restricted Fetal Rat Brain Development Which Is Associated with Increasing the Activity of PKA-CaMKII/c-fos Signal Pathway. Taurine 10-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 149-154 26322574-1 2015 This study aimed to explore whether antenatal supplement of taurine can improve the brain development of fetal rats with intrauterine growth restriction (IUGR) via protein kinase A (PKA)-Ca(2+)/calmodulin-dependent protein kinase II (CaMKII)/c-fos pathway. Taurine 60-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 242-247 26322574-4 2015 Western blot and qRT-PCR revealed that antenatal taurine supplementation significantly increased PKA, CaMKII, and c-fos protein and mRNA levels in fetal rats brain with IUGR (p < 0.05). Taurine 49-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 26322574-5 2015 Immunohistochemistry results showed that the numbers of PKA-, CaMKII-, and c-fos-positive cells were significantly decreased in the IUGR groups compared with the controls; while antenatal taurine could significantly increase the positive cell numbers (p < 0.05). Taurine 188-195 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 26322574-6 2015 So, we conclude that antenatal taurine improves IUGR fetal brain development which is associated with increasing the activity of PKA-CaMKII/c-fos signal pathway. Taurine 31-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 26189794-0 2015 Role of thalamic nuclei in the modulation of Fos expression within the cerebral cortex during hypertonic saline-induced muscle nociception. Sodium Chloride 105-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 26189794-3 2015 Following a unilateral intramuscular injection of 5.8% saline into the gastrocnemius muscle, Fos expression within the bilateral, different areas of the cerebral cortex except S2 was significantly increased (P<0.05). Sodium Chloride 55-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 26189794-4 2015 Particularly, the increases in Fos expression within the cingulate cortex and the insular cortex occurred at 0.5h, 4h and reached the peak level at 4h, 16h, respectively. 4h 115-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 26189794-4 2015 Particularly, the increases in Fos expression within the cingulate cortex and the insular cortex occurred at 0.5h, 4h and reached the peak level at 4h, 16h, respectively. 4h 148-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 26189794-5 2015 Electrolytic lesion of the contralateral thalamic MD and VM nuclei significantly blocked the 5.8% saline intramuscularly induced increases in Fos expression within the bilateral cingulate and insular cortices, respectively. Sodium Chloride 98-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-145 26189794-6 2015 Additionally, the 5.8% saline-induced Fos expression in the cingulate cortex and the insular cortex were dose-dependently attenuated by microinjection of mu-opioid antagonist beta-funaltrexamine hydrochloride into the thalamic MD and VM nuclei. Sodium Chloride 23-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 26189794-6 2015 Additionally, the 5.8% saline-induced Fos expression in the cingulate cortex and the insular cortex were dose-dependently attenuated by microinjection of mu-opioid antagonist beta-funaltrexamine hydrochloride into the thalamic MD and VM nuclei. beta-Funaltrexamine hydrochloride 175-208 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 25241022-11 2015 CONCLUSIONS: Supplementing diets with the GOS/FOS( ) mixture increased bone mineralization, density and structure due to an increase in Ca, P and Mg absorptions. Phosphorus 140-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 25241022-11 2015 CONCLUSIONS: Supplementing diets with the GOS/FOS( ) mixture increased bone mineralization, density and structure due to an increase in Ca, P and Mg absorptions. Magnesium 146-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 26330755-4 2015 Both microinjection of glutamate receptor agonists (NMDA and AMPA) into the MVN or rostral ventrolateral medullary nucleus (RVLM) and SNP-induced hypotension led to increased number of c-Fos positive neurons in the intermediolateral cell column of the middle thoracic spinal regions and increased blood epinephrine levels. Epinephrine 303-314 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 185-190 26330755-5 2015 Pretreatment with microinjection of glutamate receptor antagonists (MK-801 and CNQX) into the MVN or RVLM prevented the increased number of c-Fos positive neurons resulting from SNP-induced hypotension, and reversed the increased blood epinephrine levels. Dizocilpine Maleate 68-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 26330755-5 2015 Pretreatment with microinjection of glutamate receptor antagonists (MK-801 and CNQX) into the MVN or RVLM prevented the increased number of c-Fos positive neurons resulting from SNP-induced hypotension, and reversed the increased blood epinephrine levels. 6-Cyano-7-nitroquinoxaline-2,3-dione 79-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 26081451-8 2015 The mRNA levels of c-fos in the spinal cord and ASIC3 in the DRG in the zoledronic acid group were reduced 14 and 21 days after inoculation, and this reduction was observed in the paclitaxel group 21 days after inoculation. Zoledronic Acid 72-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 26081451-8 2015 The mRNA levels of c-fos in the spinal cord and ASIC3 in the DRG in the zoledronic acid group were reduced 14 and 21 days after inoculation, and this reduction was observed in the paclitaxel group 21 days after inoculation. Paclitaxel 180-190 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 26082062-0 2015 Estradiol suppresses neuronal firing activity and c-Fos expression in the lateral habenula. Estradiol 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 26082062-3 2015 The present study aimed to determine the effects of estradiol on LHb neurons, by observing neuronal firing activity, and c-Fos mRNA and protein expression levels in the LHb using whole cell recording, reverse transcription-quantitative polymerase chain reaction and western blotting. 5-[[[(2~{r},3~{s},4~{r},5~{r})-5-(6-Aminopurin-9-Yl)-3,4-Bis(Oxidanyl)oxolan-2-Yl]methylamino]methyl]-4-Azanyl-1-(Methoxymethyl)pyrimidin-2-One 169-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-126 26057446-0 2015 Prenatal ethanol exposure alters ethanol-induced Fos immunoreactivity and dopaminergic activity in the mesocorticolimbic pathway of the adolescent brain. Ethanol 9-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 26057446-0 2015 Prenatal ethanol exposure alters ethanol-induced Fos immunoreactivity and dopaminergic activity in the mesocorticolimbic pathway of the adolescent brain. Ethanol 33-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 26057446-9 2015 Ethanol-induced Fos-ir at AcbSh was found after 1.25g/kg and 2.5g/kg ethanol, in VE and PEE rats, respectively. Ethanol 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 26057446-9 2015 Ethanol-induced Fos-ir at AcbSh was found after 1.25g/kg and 2.5g/kg ethanol, in VE and PEE rats, respectively. acbsh 26-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 26057446-9 2015 Ethanol-induced Fos-ir at AcbSh was found after 1.25g/kg and 2.5g/kg ethanol, in VE and PEE rats, respectively. Ethanol 69-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 26057446-10 2015 PEE did not alter ethanol-induced Fos-ir at IL but reduced ethanol-induced Fos-ir at PrL. Ethanol 59-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 26062632-2 2015 Orexin fibers and terminals are present in close proximity to hindbrain catecholaminergic neurons, and fourth ventricular (4V) orexin injections that increase food intake also increase c-Fos expression in hindbrain catecholamine neurons, suggesting that orexin neurons may stimulate feeding by activating catecholamine neurons. Catecholamines 215-228 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 185-190 26062632-4 2015 We found that 4V injection of orexin-A (0.5 nmol/rat) produced widespread activation of c-Fos in hindbrain catecholamine cell groups. Catecholamines 107-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 26142830-7 2015 In in vivo studies, oral administration of rovatirelin increased both c-Fos expression in the LC and extracellular levels of noradrenaline (NA) in the medial prefrontal cortex (mPFC) of rats. rovatirelin 43-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 25862588-8 2015 injection of cyanide, reduced Fos expression in the caudal NTS, and increased Fos expression in the rostral VLM. Cyanides 13-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 25567506-10 2015 Moreover, c-Fos study shows the optical stimulation activates restricted range in midbrain: the optical stimulation affected only dPAG and its downstreams but electric stimulation activates both the upstream and downstream circuits, in which the glutamatergic neurons are largely occupied and play important role in "Stop" and "Escape" behavior controls. dpag 130-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-15 25978516-7 2015 Systemic administration of the antiglycolytic agent, 2-deoxy-D-glucose, increased food intake and c-Fos expression in orexin neurons. Deoxyglucose 53-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 25978516-10 2015 Clozapine-N-oxide injection increased food intake and c-Fos expression in PeFLH orexin neurons as well as in paraventricular hypothalamic nucleus neurons. clozapine N-oxide 0-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 26442292-7 2015 CONCLUSION: The upregulated expressions of substance P and c-fos in the spinal cord L5-S2 sections may be associated with the pain mechanism of CP/CPPS. cpps 147-151 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 26192767-7 2015 Moreover, using c-Fos staining, a strong neuronal activation was observed in the solitary tract nucleus which contains the central pattern generator of swallowing and in the area postrema after DON intravenous injection. deoxynivalenol 194-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 25886906-5 2015 Ethanol and acetate differentially activated mRNA expression for different genes, e.g., IL-6, PPARgamma, c-Fos, Egr-1, and PNPLA3 in hepatocytes. Ethanol 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 25886906-5 2015 Ethanol and acetate differentially activated mRNA expression for different genes, e.g., IL-6, PPARgamma, c-Fos, Egr-1, and PNPLA3 in hepatocytes. Acetates 12-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 26003276-6 2015 Results suggest that circadian activation of the HPA axis may affect central, but not peripheral, effect of lithium in CTA formation, and the lithium-induced c-Fos expression in brain regions may not be effective to induce CTA unless it is coupled with the HPA axis activation. Lithium 142-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 26169054-1 2015 BACKGROUND: Many studies have demonstrated that repeated injections of nicotine can produce progressive increases in locomotor activity and enhanced expression of c-fos and tyrosine hydroxylase (TH) in brain dopaminergic areas. Nicotine 71-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-168 26169054-4 2015 This study was carried out to investigate the effects of PJ on repeated nicotine-induced behavioral sensitization of locomotor activity and c-Fos and TH expression in the rat brain using immunohistochemistry. Nicotine 72-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 25637312-5 2015 Rats with 6-OHDA lesions showed increased face rubbings in the second phase when formalin was injected ipsilaterally to the lesion, and c-Fos expression in the Vc increased. Oxidopamine 10-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 25697393-0 2015 The NR2B antagonist, ifenprodil, corrects the l-DOPA-induced deficit of bilateral movement and reduces c-Fos expression in the subthalamic nucleus of hemiparkinsonian rats. ifenprodil 21-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 25697393-5 2015 Next, in order to identify the brain area influenced by L-DOPA and ifenprodil, quantitative analysis of L-DOPA-induced c-Fos immunoreactivity was performed in various brain areas of hemi-PD following administration of L-dopa with and without ifenprodil. Levodopa 104-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 25697393-6 2015 Among brain areas with robust c-Fos expression within the motor loop circuit in dopamine-depleted hemispheres, co-administered ifenprodil markedly attenuated L-DOPA-induced c-Fos expression in only the subthalamic nucleus (STN), suggesting that the STN is the primary target for the anti-parkinsonian action of NR2B antagonists. ifenprodil 127-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 25697393-6 2015 Among brain areas with robust c-Fos expression within the motor loop circuit in dopamine-depleted hemispheres, co-administered ifenprodil markedly attenuated L-DOPA-induced c-Fos expression in only the subthalamic nucleus (STN), suggesting that the STN is the primary target for the anti-parkinsonian action of NR2B antagonists. ifenprodil 127-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 173-178 25697393-6 2015 Among brain areas with robust c-Fos expression within the motor loop circuit in dopamine-depleted hemispheres, co-administered ifenprodil markedly attenuated L-DOPA-induced c-Fos expression in only the subthalamic nucleus (STN), suggesting that the STN is the primary target for the anti-parkinsonian action of NR2B antagonists. Levodopa 158-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 173-178 26059380-13 2015 Additionally, iron dextran significantly decreased c-fos and c-jun and up-regulated cellular FLICE-inhibitory protein expression levels. Iron-Dextran Complex 14-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 26063926-0 2015 Methyl supplementation attenuates cocaine-seeking behaviors and cocaine-induced c-Fos activation in a DNA methylation-dependent manner. methyl radical 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 25841323-7 2015 We also found for the first time a significant increase 2h after PD in the number of Fos-ir neurons in the brainstem nuclei that have been shown to be involved in the inhibition of SA. Deuterium 56-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 25841323-7 2015 We also found for the first time a significant increase 2h after PD in the number of Fos-ir neurons in the brainstem nuclei that have been shown to be involved in the inhibition of SA. sa 181-183 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 26170739-6 2015 Intracisternal injection of NMDA up-regulated c-Fos expression in the trigeminal neurons of the medullary dorsal horn. N-Methylaspartate 28-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 26170739-7 2015 Subcutaneous (3 U/kg) or intracisternal (1 U/kg) administration of BoNT-A significantly reduced the number of c-Fos immunoreactive neurons in the NMDA-treated rats. N-Methylaspartate 146-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 26063926-6 2015 When compared with vehicle-pretreated rats, the immediate early gene c-Fos (a marker of neuronal activation) was upregulated in the NAc and mPFC of cocaine-pretreated rats after cocaine-primed reinstatement, and chronic MET treatment blocked its induction in both regions. nac 132-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 26063926-7 2015 Cocaine-induced c-Fos expression in the NAc was associated with reduced methylation at CpG dinucleotides in the c-Fos gene promoter, effects reversed by MET treatment. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 26063926-7 2015 Cocaine-induced c-Fos expression in the NAc was associated with reduced methylation at CpG dinucleotides in the c-Fos gene promoter, effects reversed by MET treatment. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 26063926-7 2015 Cocaine-induced c-Fos expression in the NAc was associated with reduced methylation at CpG dinucleotides in the c-Fos gene promoter, effects reversed by MET treatment. nac 40-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 26063926-7 2015 Cocaine-induced c-Fos expression in the NAc was associated with reduced methylation at CpG dinucleotides in the c-Fos gene promoter, effects reversed by MET treatment. nac 40-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 26063926-7 2015 Cocaine-induced c-Fos expression in the NAc was associated with reduced methylation at CpG dinucleotides in the c-Fos gene promoter, effects reversed by MET treatment. cytidylyl-3'-5'-guanosine 87-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 26063926-7 2015 Cocaine-induced c-Fos expression in the NAc was associated with reduced methylation at CpG dinucleotides in the c-Fos gene promoter, effects reversed by MET treatment. cytidylyl-3'-5'-guanosine 87-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 25754146-7 2015 Besides, c-Fos expression in the medial nucleus of the amygdala significantly increased after testosterone treatment compared to the GDX group, while no significant difference was observed in the central and the basolateral nuclei of the amygdala in the testosterone-treated group compared to the GDX group. Testosterone 94-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 25754146-7 2015 Besides, c-Fos expression in the medial nucleus of the amygdala significantly increased after testosterone treatment compared to the GDX group, while no significant difference was observed in the central and the basolateral nuclei of the amygdala in the testosterone-treated group compared to the GDX group. GDP-alpha-D-mannuronic acid 133-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 25754146-7 2015 Besides, c-Fos expression in the medial nucleus of the amygdala significantly increased after testosterone treatment compared to the GDX group, while no significant difference was observed in the central and the basolateral nuclei of the amygdala in the testosterone-treated group compared to the GDX group. GDP-alpha-D-mannuronic acid 297-300 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 26019338-0 2015 Incubation of methamphetamine craving is associated with selective increases in expression of Bdnf and trkb, glutamate receptors, and epigenetic enzymes in cue-activated fos-expressing dorsal striatal neurons. Methamphetamine 14-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-173 26019338-9 2015 Finally, we found that DS injections of SCH23390 (C17H18ClNO), a D1-family receptor antagonist known to block cue-induced Fos induction, decreased incubated cue-induced methamphetamine seeking after prolonged withdrawal. SCH 23390 40-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 26019338-9 2015 Finally, we found that DS injections of SCH23390 (C17H18ClNO), a D1-family receptor antagonist known to block cue-induced Fos induction, decreased incubated cue-induced methamphetamine seeking after prolonged withdrawal. 2-{(E)-[(4-butylphenyl)imino]methyl}-4-chlorophenol 50-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 26019338-9 2015 Finally, we found that DS injections of SCH23390 (C17H18ClNO), a D1-family receptor antagonist known to block cue-induced Fos induction, decreased incubated cue-induced methamphetamine seeking after prolonged withdrawal. Methamphetamine 169-184 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 24712379-4 2015 Compared with those maintained on water and chow only (water group), rats trained to drink pharmacologically relevant levels of ethanol (ethanol group) showed increased neuronal activation in the PVT, specifically the aPVT but not pPVT, as indicated by c-Fos immunoreactivity. Ethanol 128-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 253-258 26007337-5 2015 It was found that higher fructose ratios engendered lower self-administration, lower Fos expression in the lateral hypothalamus/arcuate nucleus, reduced D2R and increased MOR mRNA in the dorsal striatum and nucleus accumbens core, respectively, as well as elevated omega-6 polyunsaturated fatty acids in the liver. Fructose 25-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 25746462-5 2015 Moreover, hispidulin reduced kainic acid-induced c-Fos expression and the activation of mitogen-activated protein kinases in the hippocampus. Kainic Acid 29-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 24712379-4 2015 Compared with those maintained on water and chow only (water group), rats trained to drink pharmacologically relevant levels of ethanol (ethanol group) showed increased neuronal activation in the PVT, specifically the aPVT but not pPVT, as indicated by c-Fos immunoreactivity. Ethanol 137-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 253-258 24712379-7 2015 Similarly, ethanol gavage increased double labeling of c-Fos with OX2R but not OX1R, specifically in the aPVT. Ethanol 11-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 25640291-9 2015 Surgery was associated with significantly increased c-Fos expression in the ipsilateral dorsal horn of the spinal cord, an effect attenuated by carprofen treatment. carprofen 144-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 25533534-7 2015 Induction of c-Fos expression was observed in the hypothalamic nuclei, including the medial preoptic area (MPO) and the suprachiasmatic nucleus (SCN) shell after glycine administration. Glycine 162-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 25637091-3 2015 Systemic injections of antidepressants: the tricyclic antidepressant desipramine (DMI), the selective serotonin reuptake inhibitor fluoxetine, and the NMDA-antagonist ketamine selectively increased cFos expression (a marker of neuronal activity) in the deep layers of the ventromedial PFC (vmPFC) in rats. Desipramine 69-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 198-202 25637091-3 2015 Systemic injections of antidepressants: the tricyclic antidepressant desipramine (DMI), the selective serotonin reuptake inhibitor fluoxetine, and the NMDA-antagonist ketamine selectively increased cFos expression (a marker of neuronal activity) in the deep layers of the ventromedial PFC (vmPFC) in rats. Desipramine 82-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 198-202 25637091-3 2015 Systemic injections of antidepressants: the tricyclic antidepressant desipramine (DMI), the selective serotonin reuptake inhibitor fluoxetine, and the NMDA-antagonist ketamine selectively increased cFos expression (a marker of neuronal activity) in the deep layers of the ventromedial PFC (vmPFC) in rats. N-Methylaspartate 151-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 198-202 25637091-3 2015 Systemic injections of antidepressants: the tricyclic antidepressant desipramine (DMI), the selective serotonin reuptake inhibitor fluoxetine, and the NMDA-antagonist ketamine selectively increased cFos expression (a marker of neuronal activity) in the deep layers of the ventromedial PFC (vmPFC) in rats. Ketamine 167-175 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 198-202 25529842-8 2015 RESULTS: Exposure to a cat increased c-Fos expression in TRPV1-positive neurons, mainly in the dorsal columns of the PAG, suggesting that TRPV1-expressing neurons are activated by threatening stimuli. pag 117-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 25954134-5 2015 SNP-induced hypotension increased the expression of both pERK and c-Fos protein in the NTS, which was abolished by pretreatment with glutamate receptor antagonists (MK801 or CNQX) in the MVN. Dizocilpine Maleate 165-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 25954134-5 2015 SNP-induced hypotension increased the expression of both pERK and c-Fos protein in the NTS, which was abolished by pretreatment with glutamate receptor antagonists (MK801 or CNQX) in the MVN. 6-Cyano-7-nitroquinoxaline-2,3-dione 174-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 25724236-6 2015 RESULTS: The incidence of arrhythmia was significantly higher than that of controls (75.89% versus 0%), and Fos protein expression in the PVN also increased significantly in MCAO rats; both of them could be blocked by prior application of MK-801. Dizocilpine Maleate 239-245 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 25724236-7 2015 Intracerebroventricular injection of l-glutamate induced changes in Fos protein expression and arrhythmia similar to that in the stroke, which could also be blocked by prior application of MK-801. Glutamic Acid 37-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 25724236-7 2015 Intracerebroventricular injection of l-glutamate induced changes in Fos protein expression and arrhythmia similar to that in the stroke, which could also be blocked by prior application of MK-801. Dizocilpine Maleate 189-195 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 25796089-3 2015 Our studies show that pharmacological activation of the hindbrain energy sensor AMPK by AICAR elicits estradiol (E)-dependent patterns of Fos immunolabeling of hypothalamic metabolic loci. Estradiol 102-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-141 25796089-5 2015 In both E and O rats, AICAR treatment elicited alterations in pro-opiomelanocortin, neuropeptide Y, SF-1, and orexin-A neuropeptide expression that coincided with patterns of Fos labeling of structures containing neurons that synthesize these neurotransmitters, e.g. arcuate and ventromedial nuclei and lateral hypothalamic area. AICA ribonucleotide 22-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 175-178 25557402-0 2015 Blockade of ENaCs by amiloride induces c-Fos activation of the area postrema. Amiloride 21-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 25557402-2 2015 Here, the potent ENaC blocker amiloride was injected intraperitoneally in rats and 2h later, the c-Fos activation pattern in the CVOs was studied. Amiloride 30-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 25557402-5 2015 The AP projects to FoxP2-expressing neurons in the dorsolateral pons which include the pre-locus coeruleus nucleus and external lateral part of the parabrachial nucleus; both cell groups were c-Fos activated following systemic injections of amiloride. Amiloride 241-250 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 192-197 25592423-7 2015 The spinal neuronal response to the formalin injection into the rat hind paw was also examined through immunohistochemical analysis of c-Fos protein. Formaldehyde 36-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 25592423-8 2015 Significantly increased numbers of c-Fos-immunoreactive cells were observed in laminae I-II and V-VI of the ipsilateral spinal cord to the site of the formalin injection in rats with neonatal dopamine depletion compared with vehicle-treated rats. Formaldehyde 151-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 25592423-8 2015 Significantly increased numbers of c-Fos-immunoreactive cells were observed in laminae I-II and V-VI of the ipsilateral spinal cord to the site of the formalin injection in rats with neonatal dopamine depletion compared with vehicle-treated rats. Dopamine 192-200 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 25660619-3 2015 Immunohistochemistry was performed to determine the effects of emulsified sevoflurane on formalin-induced changes of Fos-like immunoreactive (Fos-LI)-positive neurons in the spinal cord. Sevoflurane 74-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 25660619-3 2015 Immunohistochemistry was performed to determine the effects of emulsified sevoflurane on formalin-induced changes of Fos-like immunoreactive (Fos-LI)-positive neurons in the spinal cord. Sevoflurane 74-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-145 25660619-3 2015 Immunohistochemistry was performed to determine the effects of emulsified sevoflurane on formalin-induced changes of Fos-like immunoreactive (Fos-LI)-positive neurons in the spinal cord. Formaldehyde 89-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 25660619-3 2015 Immunohistochemistry was performed to determine the effects of emulsified sevoflurane on formalin-induced changes of Fos-like immunoreactive (Fos-LI)-positive neurons in the spinal cord. Formaldehyde 89-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-145 25660619-5 2015 Furthermore, Fos-LI-positive neurons were mainly found in the ipsilateral dorsal horn after the injection of formalin. Formaldehyde 109-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 25660619-6 2015 The administration of emulsified sevoflurane significantly decreased Fos-LI-labeled neurons. Sevoflurane 33-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 25660619-7 2015 Finally, intrathecal L-arginine alone did not affect the basal pain threshold, but it significantly decreased the antinociceptive response of emulsified sevoflurane against formalin injection and the suppressive effects of sevoflurane on formalin-induced Fos protein expression (P<0.05). Arginine 21-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 255-258 25814929-3 2015 Here we investigate the expression pattern and the refractory period of c-Fos in rats and monkey"s brains after stimulation with pentylenetetrazol. Pentylenetetrazole 129-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 25814929-8 2015 The refractory period after a second PTZ stimulation was also markedly different between rats and monkeys with the latter even showing a summatory effect on c-Fos expression after a second stimulation. Pentylenetetrazole 37-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-162 25755631-9 2015 Pretreatment with naloxone blocked the pheromone-induced changes in Fos expression in the magnocellular part of the paraventricular nucleus of the hypothalamus, ventrolateral periaqueductal gray, and nPGi. Naloxone 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 25445477-6 2015 Intracisternal administration of TBOA significantly increased c-Fos immunoreactivity in naive rats. benzyloxyaspartate 33-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 25445477-7 2015 In contrast, intracisternal administration of TBOA significantly decreased the up-regulation of c-Fos immunoreactivity in the medullary dorsal horn of IL-1beta- and CFA-treated rats. benzyloxyaspartate 46-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 25445477-8 2015 However, intracisternal injection of MSO blocked the up-regulation of c-Fos immunoreactivity in CFA-treated rats only. Methionine Sulfoximine 37-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 25669753-5 2015 Hippocampal neurons projecting to either PL or BA exhibited similar degrees of context-dependent Fos expression; there were more Fos-positive neurons in each area after the renewal, as opposed, to suppression of fear. pl 41-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 25669753-5 2015 Hippocampal neurons projecting to either PL or BA exhibited similar degrees of context-dependent Fos expression; there were more Fos-positive neurons in each area after the renewal, as opposed, to suppression of fear. pl 41-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-132 25669753-5 2015 Hippocampal neurons projecting to either PL or BA exhibited similar degrees of context-dependent Fos expression; there were more Fos-positive neurons in each area after the renewal, as opposed, to suppression of fear. Barium 47-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 25460109-0 2015 c-Fos induction in mesotelencephalic dopamine pathway projection targets and dorsal striatum following oral intake of sugars and fats in rats. Dopamine 37-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 25447305-0 2015 The orexin-1 receptor antagonist SB-334867 decreases anxiety-like behavior and c-Fos expression in the hypothalamus of rats exposed to cat odor. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 33-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 25447305-2 2015 In the current experiment, c-Fos expression was used to map changes in neuronal activation following SB-334867 administration in the cat odor anxiety model. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 101-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 25447305-7 2015 In rats pretreated with SB-334867, approach scores increased and c-Fos expression decreased in the PVN and PMd. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 24-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 25460109-0 2015 c-Fos induction in mesotelencephalic dopamine pathway projection targets and dorsal striatum following oral intake of sugars and fats in rats. Sugars 118-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 25417941-1 2015 Sucrose-overeating rats with decreased anorectic response to stress showed lower stress-induced activation of c-fos expression in the lateral septum (LS). Sucrose 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 25446572-0 2015 Serotonin2C receptor stimulation inhibits cocaine-induced Fos expression and DARPP-32 phosphorylation in the rat striatum independently of dopamine outflow. Cocaine 42-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 25446572-7 2015 Conversely, Ro 60-0175 inhibited cocaine-induced Fos immunoreactivity and phosphorylation of the DA and c-AMP regulated phosphoprotein of Mr 32 kDa (DARPP-32) at threonine 75 residue in the striatum. Ro 60-0175 12-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 25446572-7 2015 Conversely, Ro 60-0175 inhibited cocaine-induced Fos immunoreactivity and phosphorylation of the DA and c-AMP regulated phosphoprotein of Mr 32 kDa (DARPP-32) at threonine 75 residue in the striatum. Cocaine 33-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 25482326-7 2015 The pretreatment of rats with midazolam before the second exposure to the aversive context significantly attenuated the conditioned fear response, lowered the serum corticosterone concentration, decreased c-Fos and CRF expressions in the MeA and in the BLA, and increased CRF complex density in M1 area only in the LR group. Midazolam 30-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 205-210 25446440-7 2015 HT saline injection increased spinal Fos expression bilaterally in both the control rats and 7 days of HU rats. Sodium Chloride 3-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 25243715-0 2015 Different spatial expressions of c-Fos in the nucleus of the solitary tract following taste stimulation with sodium, potassium, and ammonium ions in rats. Sodium 109-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 25243715-0 2015 Different spatial expressions of c-Fos in the nucleus of the solitary tract following taste stimulation with sodium, potassium, and ammonium ions in rats. Potassium 117-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 25243715-0 2015 Different spatial expressions of c-Fos in the nucleus of the solitary tract following taste stimulation with sodium, potassium, and ammonium ions in rats. Ammonium Compounds 132-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 25445358-5 2015 Fos-immunoreactive nuclei were counted in rostral PAG and Edinger-Westphal nuclei (PAG-EWn bregma -6.5 mm), and caudal PAG (bregma -8 mm) of 17 adult male Sprague-Dawley rats after KCl-induced CSD under chloral hydrate anesthesia. pag 50-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 25445358-5 2015 Fos-immunoreactive nuclei were counted in rostral PAG and Edinger-Westphal nuclei (PAG-EWn bregma -6.5 mm), and caudal PAG (bregma -8 mm) of 17 adult male Sprague-Dawley rats after KCl-induced CSD under chloral hydrate anesthesia. pag 83-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 25446440-8 2015 The HT saline-induced bilateral increase of spinal Fos occurred within 0.5h and reached its peak within 1h, after which it gradually returned to the control levels within 8h. Sodium Chloride 7-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 25446440-10 2015 In superficial laminae, the HT saline-induced increases in Fos expression were higher and in the middle and deep laminae V-VI lower in the 7 days of HU than control rats. Sodium Chloride 31-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 25445358-5 2015 Fos-immunoreactive nuclei were counted in rostral PAG and Edinger-Westphal nuclei (PAG-EWn bregma -6.5 mm), and caudal PAG (bregma -8 mm) of 17 adult male Sprague-Dawley rats after KCl-induced CSD under chloral hydrate anesthesia. bregma 91-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 25451087-0 2015 Region-specific effects of isoflurane anesthesia on Fos immunoreactivity in response to intravenous cocaine challenge in rats with a history of repeated cocaine administration. Isoflurane 27-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 25445358-5 2015 Fos-immunoreactive nuclei were counted in rostral PAG and Edinger-Westphal nuclei (PAG-EWn bregma -6.5 mm), and caudal PAG (bregma -8 mm) of 17 adult male Sprague-Dawley rats after KCl-induced CSD under chloral hydrate anesthesia. Potassium Chloride 181-184 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 25445358-5 2015 Fos-immunoreactive nuclei were counted in rostral PAG and Edinger-Westphal nuclei (PAG-EWn bregma -6.5 mm), and caudal PAG (bregma -8 mm) of 17 adult male Sprague-Dawley rats after KCl-induced CSD under chloral hydrate anesthesia. Chloral Hydrate 203-218 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 25445358-7 2015 We found that the number of Fos-immunoreactive nuclei in the PAG decreased after CSD provocation. pag 61-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 25451087-12 2015 We found that challenge injections of cocaine following a regimen of repeated cocaine exposure resulted in Fos expression in the prefrontal cortex and striatum roughly equivalent to that found in rats who had received the cocaine challenge after a history of vehicle injections. Cocaine 38-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 25451087-0 2015 Region-specific effects of isoflurane anesthesia on Fos immunoreactivity in response to intravenous cocaine challenge in rats with a history of repeated cocaine administration. Cocaine 100-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 25451087-12 2015 We found that challenge injections of cocaine following a regimen of repeated cocaine exposure resulted in Fos expression in the prefrontal cortex and striatum roughly equivalent to that found in rats who had received the cocaine challenge after a history of vehicle injections. Cocaine 78-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 25451087-0 2015 Region-specific effects of isoflurane anesthesia on Fos immunoreactivity in response to intravenous cocaine challenge in rats with a history of repeated cocaine administration. Cocaine 153-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 25451087-12 2015 We found that challenge injections of cocaine following a regimen of repeated cocaine exposure resulted in Fos expression in the prefrontal cortex and striatum roughly equivalent to that found in rats who had received the cocaine challenge after a history of vehicle injections. Cocaine 78-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 25451087-13 2015 Additionally, isoflurane anesthesia resulted in a heterogeneous attenuation of cocaine-induced Fos expression, with the most robust effect in the orbital cortex but no effect in the nucleus accumbens core (NAcC). Isoflurane 14-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 25451087-2 2015 administration of cocaine increases Fos immunoreactivity in rats under isoflurane anesthesia. Cocaine 18-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-39 25451087-13 2015 Additionally, isoflurane anesthesia resulted in a heterogeneous attenuation of cocaine-induced Fos expression, with the most robust effect in the orbital cortex but no effect in the nucleus accumbens core (NAcC). Cocaine 79-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 25451087-2 2015 administration of cocaine increases Fos immunoreactivity in rats under isoflurane anesthesia. Isoflurane 71-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-39 25451087-14 2015 These results indicate that cocaine-induced Fos is preserved in the NAcC under isoflurane, suggesting that isoflurane can be used in imaging studies involving cocaine effects in this region. Cocaine 28-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 25451087-3 2015 Given that Fos expression is a marker of neural activation, the results suggested that isoflurane is appropriate for imaging cocaine effects under anesthesia. Isoflurane 87-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 25451087-14 2015 These results indicate that cocaine-induced Fos is preserved in the NAcC under isoflurane, suggesting that isoflurane can be used in imaging studies involving cocaine effects in this region. nacc 68-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 25451087-14 2015 These results indicate that cocaine-induced Fos is preserved in the NAcC under isoflurane, suggesting that isoflurane can be used in imaging studies involving cocaine effects in this region. Isoflurane 79-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 25451087-14 2015 These results indicate that cocaine-induced Fos is preserved in the NAcC under isoflurane, suggesting that isoflurane can be used in imaging studies involving cocaine effects in this region. Isoflurane 107-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 25960609-8 2015 MFB-DBS led to increased and long-lasting c-fos expression in target regions of the mesolimbic/mesocortical system. mfb-dbs 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 25149913-2 2015 In the present study, alcohol-induced adaptations in (1) anxiety-like behavior, (2) patterns of c-Fos activation and (3) subcellular distribution of corticotropin releasing factor receptor in locus coeruleus (LC) neurons was investigated in male and female Sprague-Dawley rats that were chronically exposed to ethanol using a liquid diet. Alcohols 22-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 25149913-4 2015 Ethanol-induced sex differences were observed with increased c-Fos expression in LC neurons of female ethanol-treated subjects compared to controls or male subjects. Ethanol 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 25149913-4 2015 Ethanol-induced sex differences were observed with increased c-Fos expression in LC neurons of female ethanol-treated subjects compared to controls or male subjects. Ethanol 102-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 25466986-4 2015 We detected neuronal hyperactivity through the expression of the glutamate-induced c-fos protein, by means of immunohistochemistry and immunoblotting in the vermis and in the hemispheres. Glutamic Acid 65-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 25466986-6 2015 Following the 4-aminopyridine seizures, the c-fos expression of cerebellar granule cells was significantly elevated at 1.5h in every lobule. 4-Aminopyridine 14-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 25522867-7 2015 Untreated STZ rats presented increased levels of 8-OHdG immunoreaction, higher numbers of Fos-immunoreacted neurons and high levels of co-localization of 8-OHdG and Fos in laminae I-III. Streptozocin 10-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 25522867-7 2015 Untreated STZ rats presented increased levels of 8-OHdG immunoreaction, higher numbers of Fos-immunoreacted neurons and high levels of co-localization of 8-OHdG and Fos in laminae I-III. Streptozocin 10-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-168 25327971-9 2015 Acute valproic acid administration also significantly blocked CSD-induced c-Fos expression in the TNC and TRN. Valproic Acid 6-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 25106627-6 2015 There was a significant decrease in formalin-induced biphasic behavioural response and c-Fos and nNOS immunoreactivity in the rats that were pre-treated with probenecid. Probenecid 158-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 26221181-10 2015 Furthermore, c-Fos expression was significantly facilitated in CA1 area after AMI, which was reduced by EA stimulation. amicoumacin A 78-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 26213855-10 2015 In the Fos/Hcrt co-localization assessments, ASE significantly reduced the number of Fos/Hcrt neurons in the medial, but not lateral, LH portion in ASE and CMS+ASE groups. asenapine 45-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 7-10 25553641-0 2015 Anxiety-like behaviour and c-fos expression in rats that inhaled vetiver essential oil. vetiver essential oil 65-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 26213855-10 2015 In the Fos/Hcrt co-localization assessments, ASE significantly reduced the number of Fos/Hcrt neurons in the medial, but not lateral, LH portion in ASE and CMS+ASE groups. asenapine 45-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 25553641-7 2015 VEO and diazepam significantly increased c-fos expression in the lateral division of the central amygdaloid nucleus (CeL). Diazepam 8-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 25481016-8 2014 Our findings revealed that in saline- or morphine-treated animals, acute and subcronic stress increased the p-ERK/ERK ratio, p-CREB/CREB ratio and c-fos level in the hypothalamus and hippocampus and this enhancement in morphine-treated animals, was more considerable than that in saline-treated animals. Sodium Chloride 30-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-152 25278460-10 2015 The FS-induced c-Fos immunoreactivity in the medial amygdala was different in WT and KO rats, with almost identical levels in KO and desmopressin treated animals. phenylalanylserine 4-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 25356522-3 2014 However, 2-arachidonoylglycerol (2-AG), an endocannabinoid, infused into the lateral hypothalamus of NMS rats at the beginning of the dark phase of the cycle increases rapid eye movement sleep and the expression of c-Fos on the rapid eye movement sleep promoting melanin-concentrating hormone neurons. glyceryl 2-arachidonate 9-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 215-220 25356522-3 2014 However, 2-arachidonoylglycerol (2-AG), an endocannabinoid, infused into the lateral hypothalamus of NMS rats at the beginning of the dark phase of the cycle increases rapid eye movement sleep and the expression of c-Fos on the rapid eye movement sleep promoting melanin-concentrating hormone neurons. glyceryl 2-arachidonate 33-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 215-220 25356522-3 2014 However, 2-arachidonoylglycerol (2-AG), an endocannabinoid, infused into the lateral hypothalamus of NMS rats at the beginning of the dark phase of the cycle increases rapid eye movement sleep and the expression of c-Fos on the rapid eye movement sleep promoting melanin-concentrating hormone neurons. Endocannabinoids 43-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 215-220 25481016-3 2014 In the current study, effects of acute and subchronic stress on the alteration of p-ERK, p-CREB and c-fos levels in the hypothalamus and hippocampus of saline- or morphine-treated animals during morphine-induced conditioned place preference (CPP) procedure were investigated. Sodium Chloride 152-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 25481016-3 2014 In the current study, effects of acute and subchronic stress on the alteration of p-ERK, p-CREB and c-fos levels in the hypothalamus and hippocampus of saline- or morphine-treated animals during morphine-induced conditioned place preference (CPP) procedure were investigated. Morphine 163-171 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 25481016-7 2014 The results indicated that in saline- or morphine-treated animals, p-ERK/ERK ratio, p-CREB/CREB ratio and c-fos level increased after application of acute and subchronic stress (except for p-ERK/ERK ratio in morphine-control group). Morphine 41-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 25050821-0 2014 Cocaine self-administration and extinction alter medullary noradrenergic and limbic forebrain cFos responses to acute, noncontingent cocaine injections in adult rats. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-98 25050821-0 2014 Cocaine self-administration and extinction alter medullary noradrenergic and limbic forebrain cFos responses to acute, noncontingent cocaine injections in adult rats. Cocaine 133-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-98 25050821-2 2014 The present study examined the ability of acute cocaine to induce the immediate early gene product, cFos, in NA neurons and stress-related neural circuits in rats that were cocaine-naive, or had a history of cocaine self-administration with or without extinction. Cocaine 48-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-104 25050821-9 2014 Extinction attenuated cocaine-induced cFos activation in NA neurons of the caudal ventrolateral medulla (A1/C1 cell groups), and attenuated cFos within the paraventricular nucleus of the hypothalamus, the apex of the central neuroendocrine stress axis. Cocaine 22-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-42 25050821-10 2014 Cocaine consistently increased cFos in the bed nucleus of the stria terminalis, regardless of history. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-35 24962557-5 2015 The relative number of cells expressing PV (PV+, 8-9%) did not change with age in sham-controls and also the increase in cortical c-Fos expression induced by iTBS was not principally age-dependent. itbs 158-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 25481016-8 2014 Our findings revealed that in saline- or morphine-treated animals, acute and subcronic stress increased the p-ERK/ERK ratio, p-CREB/CREB ratio and c-fos level in the hypothalamus and hippocampus and this enhancement in morphine-treated animals, was more considerable than that in saline-treated animals. Morphine 41-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-152 25454411-0 2014 Effects of differential rearing on amphetamine-induced c-fos expression in rats. Amphetamine 35-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 25454411-2 2014 Exposure to novelty and psychostimulants induces c-fos expression in neurons in the mesolimbic dopamine (DA) pathway. Dopamine 95-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 25454411-2 2014 Exposure to novelty and psychostimulants induces c-fos expression in neurons in the mesolimbic dopamine (DA) pathway. Dopamine 105-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 25454411-8 2014 Additionally, IC amphetamine rats displayed greater c-fos expression in the NAcc compared to IC saline rats, while EC saline rats displayed greater c-fos expression in the prelimbic cortex compared to EC amphetamine rats. Amphetamine 17-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 25454411-8 2014 Additionally, IC amphetamine rats displayed greater c-fos expression in the NAcc compared to IC saline rats, while EC saline rats displayed greater c-fos expression in the prelimbic cortex compared to EC amphetamine rats. nacc 76-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 25454411-8 2014 Additionally, IC amphetamine rats displayed greater c-fos expression in the NAcc compared to IC saline rats, while EC saline rats displayed greater c-fos expression in the prelimbic cortex compared to EC amphetamine rats. Sodium Chloride 118-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 25454411-9 2014 CONCLUSIONS: These results suggest regional specificity of psychostimulant-induced c-fos expression in the prelimbic/NAcc pathway that is altered in differential rearing, and influences initial c-fos activation following psychostimulant exposure. nacc 117-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 25454411-9 2014 CONCLUSIONS: These results suggest regional specificity of psychostimulant-induced c-fos expression in the prelimbic/NAcc pathway that is altered in differential rearing, and influences initial c-fos activation following psychostimulant exposure. nacc 117-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 194-199 25674230-5 2014 The optical density of c-fos protein in the lung tissues of groups of the non-acute asthma, the acute asthma and the dexamethasone intervention was positively correlated with SP concentration in the bronchoalveolar lavage fluid (r = 0.908, r = 0.967, r = 0.865), and negatively correlated with the VIP concentration in the alveolar lavage (r = -0.974, r = -0.949, r = -0.962). Dexamethasone 117-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 25327342-10 2014 HM01 increased Fos-ir cell number in the area postrema, arcuate nucleus, nucleus tractus solitarius, and lumbosacral intermediolateral column of 6-OHDA rats where 6-OHDA had a lowering effect compared to controls. Oxidopamine 163-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-18 24962407-6 2014 In contrast, the number of c-Fos-positive nuclei in the DB-treated group significantly increased 1 h after gavage. denatonium 56-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 24962407-10 2014 These results suggest that intragastric DB increases neuronal c-Fos expression in the NTS 1 h after gavage and this effect is mediated by vagal afferent fibers. denatonium 40-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 25269482-5 2014 In conscious rats, glucose infusion activated CRF-containing PVN neurons and TH-containing RVLM neurons, as indexed by c-Fos immunofluorescence. Glucose 19-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 25323389-8 2014 Expression levels of c-Fos and NGF in all central micturition areas were significantly increased following the administration of caffeine. Caffeine 129-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 25323389-9 2014 The effects on contraction pressure and time were the most potent and expression levels of c-Fos and NGF were greatest at the lowest dose of caffeine. Caffeine 141-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 25405608-5 2014 Additionally, the analysis of c-Fos expression after GAL injection in the DMH was used to investigate the potential involvement of brainstem pain control centres. Dimenhydrinate 74-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 25375852-6 2014 In cells subjected to hypoxia and glucose deprivation, dissipation of the transmembrane gradient of Na+ and K+ completely eliminated increment of Fos, Atf3, Ptgs2 and Per2 mRNAs and sharply diminished augmentation expression of Klf10, Edn1, Nr4a1 and Hes1. Glucose 34-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-149 25429177-13 2014 CONCLUSION: Both Amylin and Salmon Calcitonin inhibit formalin induced c-Fos expression in the rat lumbar spinal cord when administered intrathecally. Formaldehyde 54-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 24996127-4 2014 Our capsaicin stimulus evoked c-Fos expression in a select population of neurons within rostral trigeminal nucleus caudalis in anesthetized rats. Capsaicin 4-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 25265240-1 2014 OBJECTIVES: We examined the effects of two dopamine agonists, cabergoline and rotigotine, on tacrine-induced tremor and c-Fos expression in rats. Cabergoline 62-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 25189505-4 2014 The number of c-Fos protein-like immunoreactive (Fos-LI) neurons in the medullary dorsal horn (MDH) induced by the intraoral application of capsaicin was measured. Capsaicin 140-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 25189505-6 2014 RESULTS: Larger doses of intraoral capsaicin (1, 10 and 100mug) induced vigorous licking behaviour and c-Fos response in the MDH in a reproducible manner. Capsaicin 35-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 25189505-7 2014 The magnitudes of both behavioural activity and the c-Fos response from the 10 and 100mug doses of capsaicin were significantly greater than that by the 1mug dose. Capsaicin 99-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 25189505-8 2014 Injury to the IAN exaggerated the behavioural and c-Fos responses evoked by intraoral capsaicin. Capsaicin 86-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 25265240-1 2014 OBJECTIVES: We examined the effects of two dopamine agonists, cabergoline and rotigotine, on tacrine-induced tremor and c-Fos expression in rats. rotigotine 78-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 25265240-6 2014 The number of c-Fos-positive cells was significantly enhanced in the medial striatum, nucleus accumbens core, and nucleus accumbens shell after tacrine administration, and the enhanced expression of c-Fos in these three regions was significantly attenuated by cabergoline, while rotigotine suppressed c-Fos expression in two regions except the nucleus accumbens core. Tacrine 144-151 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 25265240-6 2014 The number of c-Fos-positive cells was significantly enhanced in the medial striatum, nucleus accumbens core, and nucleus accumbens shell after tacrine administration, and the enhanced expression of c-Fos in these three regions was significantly attenuated by cabergoline, while rotigotine suppressed c-Fos expression in two regions except the nucleus accumbens core. Cabergoline 260-271 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 25265240-6 2014 The number of c-Fos-positive cells was significantly enhanced in the medial striatum, nucleus accumbens core, and nucleus accumbens shell after tacrine administration, and the enhanced expression of c-Fos in these three regions was significantly attenuated by cabergoline, while rotigotine suppressed c-Fos expression in two regions except the nucleus accumbens core. Cabergoline 260-271 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-204 25265240-6 2014 The number of c-Fos-positive cells was significantly enhanced in the medial striatum, nucleus accumbens core, and nucleus accumbens shell after tacrine administration, and the enhanced expression of c-Fos in these three regions was significantly attenuated by cabergoline, while rotigotine suppressed c-Fos expression in two regions except the nucleus accumbens core. Cabergoline 260-271 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-204 25265240-6 2014 The number of c-Fos-positive cells was significantly enhanced in the medial striatum, nucleus accumbens core, and nucleus accumbens shell after tacrine administration, and the enhanced expression of c-Fos in these three regions was significantly attenuated by cabergoline, while rotigotine suppressed c-Fos expression in two regions except the nucleus accumbens core. rotigotine 279-289 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 25265240-6 2014 The number of c-Fos-positive cells was significantly enhanced in the medial striatum, nucleus accumbens core, and nucleus accumbens shell after tacrine administration, and the enhanced expression of c-Fos in these three regions was significantly attenuated by cabergoline, while rotigotine suppressed c-Fos expression in two regions except the nucleus accumbens core. rotigotine 279-289 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-204 25265240-6 2014 The number of c-Fos-positive cells was significantly enhanced in the medial striatum, nucleus accumbens core, and nucleus accumbens shell after tacrine administration, and the enhanced expression of c-Fos in these three regions was significantly attenuated by cabergoline, while rotigotine suppressed c-Fos expression in two regions except the nucleus accumbens core. rotigotine 279-289 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 199-204 25552429-6 2014 RESULTS: Under acute stress, ABA concentrations increased in the serum, but decreased in the hypothalamus and were accompanied by increased corticosterone in the serum and c-fos expression in the hypothalamus. Abscisic Acid 29-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-177 25386121-5 2014 The results of immunofluorescent histochemical triple-staining showed that approximately 14.2 or 18.9% of GABA-IR or MOR-IR neurons also showed MOR- or GABA-immunopositive staining in lamina II; approximately 45.2 and 36.1% of the GABA-IR and MOR-IR neurons, respectively, expressed FOS protein in their nuclei induced by injecting formalin into the left lower lip of the mouth. gamma-Aminobutyric Acid 106-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 283-286 24967534-8 2014 Additionally, pre-treating clozapine with the D1 antagonist SCH23390 caused a significant, albeit non-complete, reduction in Fos-LI, highlighting the D1 agonist property of clozapine. Clozapine 27-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-128 24967534-8 2014 Additionally, pre-treating clozapine with the D1 antagonist SCH23390 caused a significant, albeit non-complete, reduction in Fos-LI, highlighting the D1 agonist property of clozapine. SCH 23390 60-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-128 25279551-4 2014 The number of Fos-IR cells was significantly larger in M1 CFA and masseter (Mass) capsaicin applied (M1 CFA/Mass cap) rats compared with M1 veh/Mass veh rats in the contralateral Vc and Vi/Vc. Capsaicin 82-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 25306252-0 2014 7-Nitroindazole enhances c-Fos expression in spinal neurons in rats realizing operant movements. 7-nitroindazole 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 25306252-2 2014 Fos-immunoreactive neurons were visualized immunohistochemically in the C6/C7 spinal segments in the control, realized operant movements animals, and/or 7-nitroindazole (7-NI) injected rats. 7-nitroindazole 153-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 24896367-8 2014 17alpha-Estradiol down-regulated glutathione, promoter activity of the rate-limiting enzyme in glutathione production, glutamate cysteine ligase (GCL), as well as c-Fos protein levels in serum deprived cells. alfatradiol 0-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-168 24896367-9 2014 The c-Fos transcription factor normally binds to the AP-1 response element in the GCL promoter resulting in increased production of glutathione as a stress response. Glutathione 132-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 24989643-0 2014 Chronic administration of haloperidol and clozapine induces differential effects on the expression of Arc and c-Fos in rat brain. Haloperidol 26-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 24989643-0 2014 Chronic administration of haloperidol and clozapine induces differential effects on the expression of Arc and c-Fos in rat brain. Clozapine 42-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 25345960-0 2014 [Camostat mesilate, a protease inhibitor, inhibits visceral sensitivity and spinal c-fos expression in rats with acute restraint stress]. camostat 1-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 24823947-9 2014 Complementary neuroanalysis revealed that extended nicotine self-administration was associated with increased c-Fos expression in brain regions implicated in habitual control of reward seeking, including activation of the dorsolateral striatum and substantia nigra pars compacta. Nicotine 51-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 24837843-4 2014 Accordingly, gabapentin increased c-fos expression in LC neurons and noradrenaline release in the PFC in normal animals, but in SNL animals, gabapentin failed to increase c-fos expression in LC neurons projecting to the PFC and failed to increase noradrenaline release in the PFC. Gabapentin 13-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 25158105-4 2014 Using young adult female rats in a dietary-induced binge eating model (30 min access to binge food with or without 24-h calorie restriction, twice a week, for 6 weeks) we measured the neural activation by c-Fos immunoreactivity to the binge food (vegetable shortening mixed with 10% sucrose) in bingeing and non-bingeing animals under acute stress (immobilization; 1 h) or no stress conditions. Sucrose 283-290 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 205-210 25086310-6 2014 Fat, ethanol, and nicotine, but not sucrose, increased the single- and double-labeling of ENK and c-Fos-ir in precisely the same brain areas, the middle parvocellular but not lateral area of the paraventricular nucleus, central but not basolateral nucleus of the AMYG, and core but not shell of the NAc. Ethanol 5-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 25086310-6 2014 Fat, ethanol, and nicotine, but not sucrose, increased the single- and double-labeling of ENK and c-Fos-ir in precisely the same brain areas, the middle parvocellular but not lateral area of the paraventricular nucleus, central but not basolateral nucleus of the AMYG, and core but not shell of the NAc. Nicotine 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 25088910-0 2014 Regional Fos-expression induced by gamma-hydroxybutyrate (GHB): comparison with gamma-butyrolactone (GBL) and effects of co-administration of the GABAB antagonist SCH 50911 and putative GHB antagonist NCS-382. Sodium Oxybate 35-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 25031227-9 2014 Immunostaining showed a significantly elevated number of c-Fos-labeled histamine neurons in caffeine-treated rats compared with saline-treated animals. Histamine 71-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 25031227-9 2014 Immunostaining showed a significantly elevated number of c-Fos-labeled histamine neurons in caffeine-treated rats compared with saline-treated animals. Caffeine 92-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 24876059-6 2014 Furthermore, c-Fos expression in lamina I/II of the spinal dorsal horn was enhanced by thermal stimulation and the enhanced expression of c-Fos was suppressed by milnacipran. Milnacipran 162-173 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 24876059-6 2014 Furthermore, c-Fos expression in lamina I/II of the spinal dorsal horn was enhanced by thermal stimulation and the enhanced expression of c-Fos was suppressed by milnacipran. Milnacipran 162-173 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 24876059-8 2014 These results indicate that the effect of milnacipran on mechanical and thermal allodynia and c-Fos expression is elicited through the alpha2 adrenoceptor, but not alpha1 adrenoceptor, and 5-HT2 and 5-HT3 receptors; furthermore, the 5-HT1A/1B receptor is involved in mechanical allodynia, but not thermal allodynia. Milnacipran 42-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 23832598-0 2014 Fos expression induced by cocaine-conditioned cues in male and female rats. Cocaine 26-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 23832598-3 2014 Here, we used a cocaine self-administration/reinstatement model to examine neuronal activation, as determined by Fos expression, following cue-induced reinstatement of cocaine seeking in male and female rats. Cocaine 16-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-116 25429177-0 2014 Intrathecal Amylin and Salmon Calcitonin Affect Formalin Induced c-Fos Expression in the Spinal Cord of Rats. Formaldehyde 48-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 25429177-3 2014 We assessed the expression of c-Fos in response to intraplantar formalin in the lumbar regions of the spinal cord in conscious rats. Formaldehyde 64-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 25016211-7 2014 In the formalin test for examining noxious response, the intrathecal administration of [Leu(11)]-SP, but not [Leu(11)]-HK-1, reduced the number of flinchings and c-Fos-positive cells in the spinal dorsal horn following formalin injection into the plantar region of the hind paw. leu(11)]-sp 88-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 25016211-7 2014 In the formalin test for examining noxious response, the intrathecal administration of [Leu(11)]-SP, but not [Leu(11)]-HK-1, reduced the number of flinchings and c-Fos-positive cells in the spinal dorsal horn following formalin injection into the plantar region of the hind paw. Leucine 88-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 25016211-9 2014 To evaluate the involvement of HK-1 and SP in pruritic processing, the effect of [Leu(11)]-HK-1 and [Leu(11)]-SP on the induction of scratching behavior and c-Fos expression by serotonin (5-HT) and histamine was evaluated. Serotonin 177-186 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-162 25016211-10 2014 The increased induction of scratching behavior and c-Fos expression by 5-HT and histamine was markedly attenuated by pretreatment with both [Leu(11)]-HK-1 and [Leu(11)]-SP, suggesting that HK-1 and SP may be involved in pruritic processing. Histamine 80-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 25016211-10 2014 The increased induction of scratching behavior and c-Fos expression by 5-HT and histamine was markedly attenuated by pretreatment with both [Leu(11)]-HK-1 and [Leu(11)]-SP, suggesting that HK-1 and SP may be involved in pruritic processing. Leucine 141-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 25088910-0 2014 Regional Fos-expression induced by gamma-hydroxybutyrate (GHB): comparison with gamma-butyrolactone (GBL) and effects of co-administration of the GABAB antagonist SCH 50911 and putative GHB antagonist NCS-382. Sodium Oxybate 58-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 25088910-2 2014 We previously reported partial overlap in the c-Fos expression patterns produced by GHB and the GABAB agonist, baclofen in rats. Sodium Oxybate 84-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 25088910-2 2014 We previously reported partial overlap in the c-Fos expression patterns produced by GHB and the GABAB agonist, baclofen in rats. gabab 96-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 25088910-2 2014 We previously reported partial overlap in the c-Fos expression patterns produced by GHB and the GABAB agonist, baclofen in rats. Baclofen 111-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 25088910-3 2014 The present study extends these earlier findings by examining the extent to which GHB Fos expression and behavioral sedation are prevented by (2S)-(+)-5,5-dimethyl-2-morpholineacetic acid (SCH 50911), a GABAB antagonist, and NCS-382, a putative antagonist at the high-affinity GHB receptor. Sodium Oxybate 82-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 25088910-3 2014 The present study extends these earlier findings by examining the extent to which GHB Fos expression and behavioral sedation are prevented by (2S)-(+)-5,5-dimethyl-2-morpholineacetic acid (SCH 50911), a GABAB antagonist, and NCS-382, a putative antagonist at the high-affinity GHB receptor. (+)-(S)-5,5-dimethylmorpholinyl-2-acetic acid 142-187 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 25088910-4 2014 We also compare Fos expression caused by GHB and its precursor gamma-butyrolactone (GBL), which is a pro-drug for GHB but lacks the high sodium content of the parent GHB molecule. Sodium Oxybate 41-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 25088910-4 2014 We also compare Fos expression caused by GHB and its precursor gamma-butyrolactone (GBL), which is a pro-drug for GHB but lacks the high sodium content of the parent GHB molecule. 4-Butyrolactone 63-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 25088910-4 2014 We also compare Fos expression caused by GHB and its precursor gamma-butyrolactone (GBL), which is a pro-drug for GHB but lacks the high sodium content of the parent GHB molecule. 4-Butyrolactone 84-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 25088910-5 2014 Both GHB (1,000 mg/kg) and GBL (600 mg/kg) induced rapid sedation in rats that lasted over 90 min and caused similar Fos expression patterns, albeit with GBL causing greater activation of the nucleus accumbens (core and shell) and dentate gyrus (granular layer). Sodium Oxybate 5-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 25088910-5 2014 Both GHB (1,000 mg/kg) and GBL (600 mg/kg) induced rapid sedation in rats that lasted over 90 min and caused similar Fos expression patterns, albeit with GBL causing greater activation of the nucleus accumbens (core and shell) and dentate gyrus (granular layer). 4-Butyrolactone 27-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-120 25088910-6 2014 Pretreatment with SCH 50911 (100mg/kg) partly reversed the sedative effects of GHB and significantly reduced GHB-induced Fos expression in only four regions: the tenia tecta, lateral habenula, dorsal raphe and laterodorsal tegmental nucleus. Sodium Oxybate 109-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-124 25088910-10 2014 Overall, this study shows a surprising lack of reversal of GHB-induced Fos expression by two relevant antagonists, both of which have marked intrinsic actions. Sodium Oxybate 59-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 25171958-0 2014 Effect of acute asenapine treatment on Fos expression in the forebrain structures under normal conditions and mild stress preconditioning in the rat. asenapine 16-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 25171958-3 2014 The aim of the present study was: (1) to delineate the quantitative and qualitative profiles of the ASE effect on Fos expression in the striatum, septum, nucleus accumbens, and the prefrontal cortex and (2) to find out whether a chronic unpredictable variable mild stress (CMS) preconditioning may modify the effect of acute ASE treatment. asenapine 100-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 25171958-15 2014 The present paper demonstrates for the first time the character of the Fos distribution in the forebrain structures induced by acute ASE treatment as well as ASE response to 21 days CMS preconditioning. asenapine 133-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 25150425-12 2014 c-Fos was mainly observed in the raphe pallidus area with significant differences between groups A and B at 3 and 6 hours after injection of CFA (P = .027 and .022, respectively). 3-chloro-4-fluoroaniline 141-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 24713611-5 2014 In the accumbens, we found reduced mGluR5 expression (immunohistochemistry and Western blot) and decreased neural activation (as measured by c-Fos immunohistochemistry) in response to a moderate alcohol dose (1 g/kg) following CORT exposure (7 days). Alcohols 195-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 24933337-0 2014 The effects of the estrus cycle and citalopram on anxiety-like behaviors and c-fos expression in rats. Citalopram 36-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 24933337-11 2014 C-fos expression at the amygdala (p=0.013) and the paraventricular thalamic nucleus (p=0.014) was significantly inhibited in the citalopram group. Citalopram 129-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 25017242-2 2014 It is known that peripheral injection of CCK increases c-Fos-immunoreactivity (Fos-IR) in the nucleus of the solitary tract (NTS) in rats, and injection of the serotonin antagonist ondansetron decreases the number of c-Fos-IR cells in the NTS. Ondansetron 181-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 25017242-2 2014 It is known that peripheral injection of CCK increases c-Fos-immunoreactivity (Fos-IR) in the nucleus of the solitary tract (NTS) in rats, and injection of the serotonin antagonist ondansetron decreases the number of c-Fos-IR cells in the NTS. Ondansetron 181-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 217-222 24802360-8 2014 OEA and found that peripheral OEA strongly activates c-fos expression in the AP, NST and in the hypothalamus of both chow and HFD fed rats. oleoylethanolamide 30-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 25571636-0 2014 [Effects of citalopram on the expression of PCNA and C-fos and cell apoptosis in rat frontal cortical neurons after stress]. Citalopram 12-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 25571636-1 2014 OBJECTIVE: To study the effects of citalopram on the expression of proliferating cell nuclear antigen (PCNA) and proto-oncogene protein (C-fos) and cell apoptosis in frontal cortical neurons of rat after stress. Citalopram 35-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 25571636-9 2014 CONCLUSION: Citalopram significantly antagonize PCNA, C-fos protein expression and cell apoptosis of rat prefrontal cortical neurons caused by chronic stress, which might be the one of mechanisms of citalopram for prevention and treatment of psychosis caused by chronic stress. Citalopram 12-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 24090794-5 2014 Immunohistochemistry was used to identify sex-dependent expression of U-50488-induced c-Fos in brain. 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer 70-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 24090794-9 2014 Sex differences in U-50488-induced c-Fos activation were observed in corticotropin releasing factor-containing neurons of the paraventricular nucleus of the hypothalamus and primarily in non-corticotropin releasing factor-containing neurons of the bed nucleus of the stria terminalis. 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer 19-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 25100956-9 2014 Restraint stress stimulated Fos-protein expression in perifornical area orexin cells, the paraventricular hypothalamic nucleus, and paraventricular thalamic nuclei, but this neuronal response was dampened in male and female rats exposed to ELS. N-[(2S,3S,4R)-3,4-dihydroxy-8-oxo-8-[(4-pentylphenyl)amino]-1-{[(2S,3R,4S,5R,6R)-3,4,5-trihydroxy-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yl]oxy}octan-2-yl]hexacosanamide 240-243 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 25234442-8 2014 L-NAME-treatment significantly reduced the exercise-induced c-Fos expression in the PVN, whereas it had no effect in the SON. NG-Nitroarginine Methyl Ester 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 24727435-5 2014 Fos expression was suppressed by 65-92% in capsaicin-treated animals, as was epinephrine (74%), norepinephrine (33%), and glucagon (47%). Capsaicin 43-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 24907695-10 2014 Acute, but not chronic, caffeine exposure elevated c-Fos expression in the NAc. Caffeine 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 24923763-0 2014 Effects of electroacupuncture on ethanol-induced impairments of spatial learning and memory and Fos expression in the hippocampus in rats. Ethanol 33-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-99 24923763-9 2014 Animals administered with ethanol emitted significantly fewer Fos expression in the hippocampal CA1 area. Ethanol 26-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 24675157-4 2014 We showed that CS(+) presentation after conditioning increased Fos expression in the basolateral nucleus of amygdala (BLA). Cesium 15-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 24797330-0 2014 Distribution of Fos-immunoreactive cells in rat forebrain and midbrain following social defeat stress and diazepam treatment. Diazepam 106-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 24829502-2 2014 We hypothesized that elevated corticosterone (Cort) within the dorsal hindbrain (DHB) would: 1) enhance arterial pressure and neuroendocrine responses to novel and repeated restraint stress, 2) increase c-Fos expression in regions of the brain involved in sympathetic stimulation during stress, and 3) recruit a vasopressin-mediated blood pressure response to acute stress. Corticosterone 30-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 203-208 24829502-2 2014 We hypothesized that elevated corticosterone (Cort) within the dorsal hindbrain (DHB) would: 1) enhance arterial pressure and neuroendocrine responses to novel and repeated restraint stress, 2) increase c-Fos expression in regions of the brain involved in sympathetic stimulation during stress, and 3) recruit a vasopressin-mediated blood pressure response to acute stress. dihydrodibutylstilbestrol 81-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 203-208 25025380-9 2014 Compared to oxycodone self-administering control rats immunized with the carrier alone, rats vaccinated with the OXY-KLH immunogen showed increased levels of adenylate cyclase 5 (Adcy5) and decreased levels of early growth response protein 2 (Egr2) and the early immediate gene c-Fos in the striatum. oxy-klh 113-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 278-283 24797330-8 2014 The diazepam treatment significantly reduced the stress-induced Fos expression in many brain regions including the prefrontal, sensory and motor cortices, septum, medial amygdaloid nucleus, medial and lateral preoptic areas, parvicellular paraventricular hypothalamic nucleus, dorsomedial hypothalamus, perifornical nucleus, tuberomammillary nucleus, association, midline and intralaminar thalami, and median and dorsal raphe nuclei. Diazepam 4-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 24797330-9 2014 In contrast, diazepam increased Fos-IR cells in the central amygdaloid nucleus, medial habenular nucleus, ventromedial hypothalamic nucleus and magnocellular lateral hypothalamus. Diazepam 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 25009258-6 2014 In the paraventricular nuclei (PVN) of the hypothalamus, DOC pretreatment diminished both AngII-induced cFos induction and neurosecretion of oxytocin, a peptide expressed in the PVN. Desoxycorticosterone 57-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-108 24113864-7 2014 In addition, the noxious thermal stimulus-induced c-Fos expression in the ventrolateral periaqueductal gray matter was significantly suppressed by concomitant ketamine and morphine. Ketamine 159-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 23425888-3 2014 High-frequency stimulation of the perforant pathway in urethane-anesthetized rats elicited expression of the immediate early genes c-fos, Arc, zif268 and pCREB133 in almost 100% of mature, calbindin-positive granule cells. Urethane 55-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-136 23425888-7 2014 Labeling with bromodeoxyuridine revealed cell age dependence of stimulation-induced c-fos, Arc and zif268 expression, with only a few cells reacting at 21 days, but with up to 75% of cells activated at 35-77 days of cell age. Bromodeoxyuridine 14-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 24335852-10 2014 Moreover, CSD and environmental stress increased Fos expression in the TNC of sumatriptan-treated rats, and these effects were blocked by topiramate. Sumatriptan 78-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 24335852-10 2014 Moreover, CSD and environmental stress increased Fos expression in the TNC of sumatriptan-treated rats, and these effects were blocked by topiramate. Topiramate 138-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 24862585-7 2014 ES, but not yoked IS, produced a large increase of Fos activity in the DMS. Einsteinium 0-2 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 24862585-8 2014 Consistent with the Fos data, D-AP5 in the DMS, but not in the DLS, prevented the inhibition of dorsal raphe nucleus 5-HT release normally produced by ES. 2-amino-4-oxo-5-phosphonopentanoic acid 30-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 24553733-7 2014 Juvenile rats infused with picrotoxin into the prelimbic mPFC and exposed to a threatening stimulus froze less, spent less time far from the threat, and increased Fos expression. Picrotoxin 27-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 24845185-7 2014 Following CVS, basal and 15 min post-restraint plasma corticosterone levels were increased by NTS GR antagonism, which was associated with an increase in Fos immunoreactivity within the PVN. Corticosterone 54-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-157 24835545-9 2014 Results showed that in the Play-only experiment exposure to the CS+ induced more FOS-IR in the medial prefrontal cortex, orbitofrontal cortex, dorsal striatum, and ventral tegmental area as compared to females from the unpaired group. Cesium 64-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-84 24631866-8 2014 5TG modified patterns of steroid feedback-associated Fos staining of A2, but not other medullary catecholamine cell groups. 5-thio-D-glucose 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 24631866-8 2014 5TG modified patterns of steroid feedback-associated Fos staining of A2, but not other medullary catecholamine cell groups. Steroids 25-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 24631866-9 2014 Intra-preoptic administration of the alpha1-adrenergic receptor agonist, methoxamine, elicited site-specific reversal of hindbrain glucoprivic suppression of gonadotropin-releasing hormone (GnRH) neuron Fos labeling and LH release. Methoxamine 73-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 203-206 24641259-0 2014 Amitriptyline up-regulates connexin43-gap junction in rat cultured cortical astrocytes via activation of the p38 and c-Fos/AP-1 signalling pathway. Amitriptyline 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 24641259-10 2014 The translocation of c-Fos from the cytosol and the nucleus of cortical astrocytes was increased by amitriptyline, and this response was dependent on p38 activity. Amitriptyline 100-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 24992726-9 2014 Moreover, by day 14, Fos-IR associated with PRE084 infusion was significantly reversed by NMDA receptor antagonist MK801, rather than BD1047. Dizocilpine Maleate 115-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 24385076-6 2014 KYNAa pre-treatment had dose-dependent, mitigating action on nitroglycerine-induced decrease in calcitonin gene-related peptide and increase in c-Fos, neuronal nitric oxide synthase and calmodulin-dependent protein kinase II alpha expression in the C1-C2. Nitroglycerin 61-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-149 24113864-7 2014 In addition, the noxious thermal stimulus-induced c-Fos expression in the ventrolateral periaqueductal gray matter was significantly suppressed by concomitant ketamine and morphine. Morphine 172-180 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 24878577-0 2014 Altered formalin-induced pain and Fos induction in the periaqueductal grey of preadolescent rats following neonatal LPS exposure. Formaldehyde 8-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 24522554-9 2014 Treatment with DMU-212 alone increased the constitutive AP-1 subunits c-Jun and c-Fos levels and c-Jun binding to TRE consensus site. dmu-212 15-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 24872549-4 2014 On test day, reexposure to the cocaine-associated Context A reinstated cocaine seeking and increased expression of the neural activity marker Fos in 3.3% of accumbens shell and 1.6% of accumbens core neurons. Cocaine 31-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-145 24872549-6 2014 We trained c-fos-lacZ transgenic rats to self-administer cocaine in Context A and extinguished their lever-pressing in Context B. Cocaine 57-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 24872549-8 2014 On test day, 3 d after induction day, the ability of Context A to reinstate cocaine seeking and increase neuronal activity in accumbens shell was attenuated when Daun02 was previously injected after exposure to Context A. Daun02 injections after exposure to the novel Context C had no effect on context-induced reinstatement of cocaine seeking despite much greater numbers of Fos-expressing neurons induced by Context C. Daun02 injections in accumbens core had no effect. Daun02 162-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 376-379 24878577-4 2014 This LPS-induced hyperalgesia was accompanied by distinct recruitment of supra-spinal regions involved in analgesia as indicated by significantly attenuated Fos-protein induction in the rostral dorsal periaqueductal grey (DPAG) as well as rostral and caudal axes of the ventrolateral PAG (VLPAG). dpag 222-226 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-160 24878577-5 2014 Formalin injections were associated with increased Fos-protein labelling in lateral habenula (LHb) as compared to medial habenula (MHb), however the intensity of this labelling did not differ as a result of neonatal immune challenge. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 24598462-2 2014 Tryptophan hydroxylase (Tryp-OH, synthetic enzyme-producing 5-HT) immunoreactive neurons in the AP of rats become c-Fos-activated following conditions in which plasma sodium levels are elevated; these include intraperitoneal injections of hypertonic saline and sodium repletion. Sodium 167-173 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 24582760-1 2014 Lithium chloride at doses sufficient to induce conditioned taste aversion (CTA) causes c-Fos expression in the paraventricular nucleus and increases the plasma level of corticosterone with activation of the hypothalamic-pituitary-adrenal axis. Lithium Chloride 0-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 24835545-2 2014 Herein, we examined the expression of FOS immunoreactivity (FOS-IR) following exposure to the odor paired with juvenile play (CS+). Cesium 126-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 24835545-2 2014 Herein, we examined the expression of FOS immunoreactivity (FOS-IR) following exposure to the odor paired with juvenile play (CS+). Cesium 126-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 24598462-2 2014 Tryptophan hydroxylase (Tryp-OH, synthetic enzyme-producing 5-HT) immunoreactive neurons in the AP of rats become c-Fos-activated following conditions in which plasma sodium levels are elevated; these include intraperitoneal injections of hypertonic saline and sodium repletion. Sodium Chloride 250-256 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 24573182-6 2014 In contrast, phenylephrine (PE)-induced rise in MAP evoked markedly attenuated bradycardia with dramatically reduced c-Fos expression in the nucleus tractus solitarius (NTS) of adult OZR compared with LZR. Phenylephrine 13-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 24573182-6 2014 In contrast, phenylephrine (PE)-induced rise in MAP evoked markedly attenuated bradycardia with dramatically reduced c-Fos expression in the nucleus tractus solitarius (NTS) of adult OZR compared with LZR. Phenylephrine 28-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 24598462-2 2014 Tryptophan hydroxylase (Tryp-OH, synthetic enzyme-producing 5-HT) immunoreactive neurons in the AP of rats become c-Fos-activated following conditions in which plasma sodium levels are elevated; these include intraperitoneal injections of hypertonic saline and sodium repletion. Sodium 261-267 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 24573182-7 2014 However, in juvenile rats, PE-induced hypertension evoked comparable bradycardia in OZR and LZR with similar or augmented c-Fos expression in NTS of the OZR. Phenylephrine 27-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 24708580-4 2014 In the present study, we intend to study the efficacy of antidotes comprising of HI-6 (1-[[[4-(aminocarbonyl)-pyridinio]-methoxy]-methyl]-2-[(hydroxyimino) methyl] pyridinium dichloride), atropine and midazolam on soman induced neurodegeneration and the expression of c-Fos, Calpain, and Bax levels in discrete rat brain areas. asoxime chloride 81-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 268-273 24617700-5 2014 In contrast, neurons in this RVLM region, including catecholamine-synthesizing neurons, did express c-Fos following induced hypotension, which reflexly activates RVLM sympathetic premotor neurons. Catecholamines 52-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 24604295-0 2014 beta-glucan reduces exercise-induced stress through downregulation of c-Fos and c-Jun expression in the brains of exhausted rats. beta-Glucans 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 24604295-4 2014 In the present study, the effects of beta-glucan on the expression of the oncogenes c-Fos and c-Jun in the hypothalamus, dentate gyrus and dorsal raphe in rats following exhaustive treadmill running were investigated. beta-Glucans 37-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 24604295-10 2014 Administration of beta-glucan resulted in an increase in the time to exhaustion and the suppression of the exercise-induced increment in c-Fos and c-Jun expression. beta-Glucans 18-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 24604295-11 2014 In conclusion, beta-glucan may exert an alleviating effect on exercise-induced stress through the suppression of c-Fos and c-Jun expression in the brains of exhausted rats. beta-Glucans 15-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 25244785-9 2014 CONCLUSION: The method of targeted damaging CSF-contacting nucleus by cholera toxin subnit B conjugated with saporin(CB-SAP) is scientific and reliable, and it results in the changes of pain threshold and expression of 5-HT and c-Fos in spinal dorsal horn of rats. cb-sap 117-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 228-233 24605826-9 2014 Estradiol-17beta treatment reduced c-Fos labeling in EGFP-GnIH neurons in the DMN of young ovariectomized adult females but had no effect in middle-aged females. Estradiol 0-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 24676683-3 2014 The administration of both 2DG and insulin stimulated food intake and induced c-Fos expression in the ORX-A neurons corresponding to food intake, but not in the MCH neurons. Deoxyglucose 27-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 24818491-17 2014 CONCLUSION: Deep EA stimulation of GB 30 can improve the pathological changes and function of the injured sciatic nerve in the rat, which is closely associated with its effects in up-regulating NGF expression and down-regulating Fos expression. gb 30 35-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 229-232 24507935-6 2014 Ondansetron injection into the MS/vDB increases Fos-IR in different brain areas including the limbic system (central amygdala and ventral part of the bed nucleus of the stria terminalis), hypothalamus (medial parvocellular parts of the paraventricular nucleus, anterodorsal preoptic area, dorsomedial hypothalamic nucleus), mesencephalon (ventrolateral periaqueductal gray region) and rhombencephalon (lateral parabrachial nucleus) in sham rats. Ondansetron 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 24492838-10 2014 Intracerebroventricular administration of the ERK phosphorylation inhibitor U 0126 before a hyperosmotic challenge reduced the number of both phosphoERK-immunopositive neurones and Fos expressing neurones in osmosensitive forebrain regions. U 0126 76-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 181-184 24530741-8 2014 7-Nitroindazole, a selective nNOS inhibitor, abolished Fos-immunoreactivity induced by bombesin in COX-1-immunoreactive FG-labeled PVN neurons. 7-nitroindazole 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 24584054-3 2014 We found that projections to VTA from the rostral ventral pallidum (RVP), but not the caudal ventral pallidum (CVP), were robustly Fos activated during cue-induced reinstatement of cocaine seeking--a rat model of relapse in addiction. Cocaine 181-188 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-134 24412800-7 2014 In addition, acupuncture at ST36 and/or upper lip formalin induced c-Fos expression in the nucleus accumbens and in rostral ventral medulla. Formaldehyde 50-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 24212060-3 2014 In a further experiment, we compared the effects of two different doses of NaBu (200 and 1200 mg/kg) and equimolar saline solutions on peripheral neuroendocrine markers and brain c-Fos expression to demonstrate a specific stress-like effect of NaBu that is not related to hypertonicity and to localise putatively involved brain areas. sethoxydim 75-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-184 24212060-5 2014 The equimolar (hypertonic) saline solution also activated the HPA axis and the c-Fos expression in the paraventricular nucleus of the hypothalamus (PVN), a key area for the control of the HPA axis, but the effects were of a lower magnitude than those of NaBu. Sodium Chloride 27-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 24212060-7 2014 However, only the latter area of the NaBu group showed enhanced c-Fos expression that was significantly higher than that after hypertonic saline. sethoxydim 37-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 24447511-9 2014 Inhibition of P2X7R by BBG or small-interference RNA targeting P2X7 in the RVM markedly reduced 5-HT level and Fos expression in the spinal cord. coomassie Brilliant Blue 23-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 24468031-9 2014 Rats in the OVx group consistently consumed significantly less capsaicin and exhibited significantly higher counts of capsaicin-evoked Fos-like immunoreactivity in the dorsomedial trigeminal subnucleus caudalis (Vc) compared to all other treatment groups. Capsaicin 118-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-138 24723855-4 2014 Atropine blocked c-Fos expression in the cortex and BG, despite high c-Fos expression in the sub-cortical arousal neuronal groups and thalamus, indicating that cortical activity is required for BG activation. Atropine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 24486893-4 2014 Expression of c-Fos protein increased significantly in the NTS in the sham group after sodium nitroprusside (SNP) administration. Nitroprusside 87-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 24658263-9 2014 Dex or Ropi displayed a short-term analgesia in a dose-dependent manner, and consecutive daily administrations of their combination showed synergistic analgesia and remarkably down-regulated neuronal and astrocytic activations indicated by decreases in the number of Fos-ir neurons and the GFAP expression within the SDH, respectively. Dexmedetomidine 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 267-270 24342748-0 2014 Chronic wheel running affects cocaine-induced c-Fos expression in brain reward areas in rats. Cocaine 30-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 24658263-9 2014 Dex or Ropi displayed a short-term analgesia in a dose-dependent manner, and consecutive daily administrations of their combination showed synergistic analgesia and remarkably down-regulated neuronal and astrocytic activations indicated by decreases in the number of Fos-ir neurons and the GFAP expression within the SDH, respectively. Ropivacaine 7-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 267-270 24342748-7 2014 The c-Fos transcription factor is a measure of cellular activity and was used to quantify cocaine-induced activation of reward-processing areas of the brain: nucleus accumbens (NAc), caudate putamen (CPu), medial prefrontal cortex (mPFC), and orbitofrontal cortex (OFC). Cocaine 90-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 24342748-8 2014 The mean fold change in cocaine-induced c-Fos cell counts relative to saline-induced c-Fos cell counts was significantly higher in exercising compared to control rats in the NAc core, dorsomedial and dorsolateral CPu, the prelimbic area, and the OFC, indicating differential cocaine-specific cellular activation of brain reward circuitry between exercising and control animals. Cocaine 24-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 24342748-8 2014 The mean fold change in cocaine-induced c-Fos cell counts relative to saline-induced c-Fos cell counts was significantly higher in exercising compared to control rats in the NAc core, dorsomedial and dorsolateral CPu, the prelimbic area, and the OFC, indicating differential cocaine-specific cellular activation of brain reward circuitry between exercising and control animals. Sodium Chloride 70-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 24342748-8 2014 The mean fold change in cocaine-induced c-Fos cell counts relative to saline-induced c-Fos cell counts was significantly higher in exercising compared to control rats in the NAc core, dorsomedial and dorsolateral CPu, the prelimbic area, and the OFC, indicating differential cocaine-specific cellular activation of brain reward circuitry between exercising and control animals. Cocaine 275-282 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 24381176-7 2014 In rats with chronic low or high sodium intakes, the low-sodium diet was associated with significantly higher plasma aldosterone, MR mRNA and protein expression, and c-Fos immunoreactivity within labeled preautonomic neurons. Sodium 57-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-171 24292370-0 2014 Changes in the levels of p-ERK, p-CREB, and c-fos in rat mesocorticolimbic dopaminergic system after morphine-induced conditioned place preference: the role of acute and subchronic stress. Morphine 101-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 24292370-12 2014 Our findings suggest that in saline- or morphine-treated animals, acute and subchronic stress increases p-ERK, p-CREB, and c-fos levels in the mesocorticolimbic system. Sodium Chloride 29-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 24292370-12 2014 Our findings suggest that in saline- or morphine-treated animals, acute and subchronic stress increases p-ERK, p-CREB, and c-fos levels in the mesocorticolimbic system. Morphine 40-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 24342046-9 2014 Curcumin treatment greatly decreased the levels of c-Jun, c-Fos, SphK1, and FN in the kidney tissues of diabetic rats. Curcumin 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 24304472-8 2014 Treatment with U0126 reversed the effects of NE on c-Fos expression, p38 mitogen-activated protein kinase (MAPK) phosphorylation and TNF-alpha production, but not NF-kappaB activation in LPS-challenged cardiomyocytes. U 0126 15-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 24247001-6 2014 Furthermore, intrathecal administration of HK-1 (1-5) and SP (1-5) markedly attenuated the induction of flinching and enhancement of c-Fos expression in the spinal cord following the intradermal administration of formalin, a noxious stimulant, while pretreatment with HK-1 (1-5), but not SP (1-5), markedly attenuated the induction of scratching behavior by subcutaneous administration of pruritic agents, such as serotonin or histamine. Formaldehyde 213-221 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 24291670-9 2014 Quantitative image analysis showed that the numbers of c-Fos-immunoreactive neurons in the left AIC and PIC were significantly lower in the FS+CFA group (L AIC, 95.9+-6.8; L PIC, 181.9+-23.1) than those in the naive group (L AIC, 151.1+-19.3, p<0.05; L PIC, 274.2+-37.3, p<0.05). phenylalanylserine 140-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 24118230-11 2014 A significant c-FOS activation occurred after one hour of Hip-DBS in the ipsilateral amygdala; there was no contralateral amygdala activation, and by six hours, no amygdala activation was noted. hip-dbs 58-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 24460246-7 2014 Analysis of certain transcription factors revealed that nuclear expression of c-Fos and the mRNA expression were suppressed by EPA and DHA. Eicosapentaenoic Acid 127-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 24460246-7 2014 Analysis of certain transcription factors revealed that nuclear expression of c-Fos and the mRNA expression were suppressed by EPA and DHA. Docosahexaenoic Acids 135-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 24157491-7 2014 Nicotine pretreatment enhanced quinpirole-induced c-fos mRNA expression in the hypothalamic paraventricular and supraoptic nuclei in adolescents only. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 24231720-4 2014 Luminally applied estradiol induced a rapid and significant decrease in the visceromotor response to colorectal distension, with increased c-Fos expression in nodose ganglion neurons. Estradiol 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 24157491-7 2014 Nicotine pretreatment enhanced quinpirole-induced c-fos mRNA expression in the hypothalamic paraventricular and supraoptic nuclei in adolescents only. Quinpirole 31-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 24157491-8 2014 Adolescent nicotine pretreatment enhanced c-fos mRNA expression in corticotropin releasing factor (CRF) cells of the paraventricular nucleus, and enhancement of penile erection was blocked by the CRF-1 receptor antagonist, CP 376,396. Nicotine 11-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 24333564-0 2014 Nitric oxide in the commissural nucleus tractus solitarii regulates carotid chemoreception hyperglycemic reflex and c-Fos expression. Nitric Oxide 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 24186846-0 2014 N-(2-hydroxy phenyl) acetamide produces profound inhibition of c-Fos protein and mRNA expression in the brain of adjuvant-induced arthritic rats. 2-hydroxyacetanilide 0-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 24186846-10 2014 In comparison to the control group, the treatment of NA-2 treatment was found to block the development of the arthritis-induced c-Fos protein and mRNA expression and peripheral edema. sodium sulfide 53-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-133 24304007-0 2014 The antagonism of aluminum against fluoride-induced oxidative stress and c-Fos overexpression in rat testes. Aluminum 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 24304007-3 2014 The present study aims to define a possible mechanism of NaF-induced reproductive toxicity with respect to mineral, oxidative stress and c-Fos expression and the role of aluminum (Al) in intervening the toxic effect of NaF on rat testes. Sodium Fluoride 57-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 24304007-8 2014 Meanwhile, the protein expression of c-Fos increased significantly in the 1.0 and 2.0 mg NaF groups compared with the control group. Sodium Fluoride 89-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 24304007-10 2014 The present study suggested that NaF could decrease the contents of Ca, Fe and Mg and enhance oxidative stress leading to c-Fos overexpression, and some deleterious effects were more prominent at lower NaF intake. Sodium Fluoride 33-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 24316406-6 2014 Intrathecal administration of Homer 1b/c antisense oligonucleotides not only markedly reduced the expression of Homer 1b/c protein, but also attenuated CFA-induced inflammation, spinal CREB phosphorylation, and Fos expression. Oligonucleotides 51-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 211-214 24465697-2 2014 We report here the effects of the prebiotic polysaccharide inulin and its hydrolysed form FOS on this bacterium. Polysaccharides 44-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 24377428-7 2014 Immunochemical induction of c-fos in the cortex was significantly suppressed (p < 0.01) after administration of xenon. Xenon 115-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 24377428-8 2014 The molecular analysis revealed significant effects of N2O and xenon administration on c-fos and MMP-9 expression. Nitrous Oxide 55-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 24377428-8 2014 The molecular analysis revealed significant effects of N2O and xenon administration on c-fos and MMP-9 expression. Xenon 63-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 24000375-5 2014 The effect of olcegepant, L-nitro-arginine methyl ester (L-NAME) and neurokinin (NK)-1 receptor antagonist L-733060 were analysed on Fos activation. 3-((3,5-bis(trifluoromethyl)phenyl)methyloxy)-2-phenylpiperidine 107-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-136 24000375-9 2014 Pre-treatment with L-NAME and L-733060 also significantly inhibited GTN induced Fos expression. NG-Nitroarginine Methyl Ester 19-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 24000375-9 2014 Pre-treatment with L-NAME and L-733060 also significantly inhibited GTN induced Fos expression. 3-((3,5-bis(trifluoromethyl)phenyl)methyloxy)-2-phenylpiperidine 30-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 24000375-9 2014 Pre-treatment with L-NAME and L-733060 also significantly inhibited GTN induced Fos expression. Nitroglycerin 68-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 24000375-10 2014 CONCLUSION: The present study indicates that blockers of CGRP, NOS and NK-1 receptors all inhibit GTN induced Fos activation. Nitroglycerin 98-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-113 24333477-8 2014 Finally, we examined the effects of an antagonist(s) that reduced tranexamic acid-induced kaolin intake on the increase in c-Fos immunoreactive cells. Tranexamic Acid 66-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 24333477-11 2014 Tranexamic acid significantly increased c-Fos immunoreactive cells by approximately 5.5-fold and 22-fold in the area postrema and nucleus of solitary tract, respectively. Tranexamic Acid 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 24409327-9 2014 However, the immunohistochemical analysis revealed that LPA 18:1 increased c-Fos expression in the DPAG. dpag 99-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 25530786-10 2014 Both EA and morphine could equally inhibit PI-induced p-ERK and Fos inductions. Morphine 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 24140441-0 2014 Effect of rhynchophylline on the expression of p-CREB and sc-Fos in triatum and hippocampal CA1 area of methamphetamine-induced conditioned place preference rats. rhyncophylline 10-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 24140441-0 2014 Effect of rhynchophylline on the expression of p-CREB and sc-Fos in triatum and hippocampal CA1 area of methamphetamine-induced conditioned place preference rats. Methamphetamine 104-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 24140441-1 2014 To explore the effect of rhynchophylline (Rhy) on the expression of p-CREB and c-Fos in the striatum and hippocampal CA1 area of methamphetamine-induced conditioned place preference (CPP) rat, methamphetamine (2 mg/kg) was injected to rats and the conditioned place preference was observed in these rats treated with or without Rhy. rhyncophylline 25-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 24140441-1 2014 To explore the effect of rhynchophylline (Rhy) on the expression of p-CREB and c-Fos in the striatum and hippocampal CA1 area of methamphetamine-induced conditioned place preference (CPP) rat, methamphetamine (2 mg/kg) was injected to rats and the conditioned place preference was observed in these rats treated with or without Rhy. Methamphetamine 129-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 24140441-4 2014 Methamphetamine also increased the number of p-CREB positive cells in the striatum and hippocampal CA1 zone, as well as p-Fos positive cells. Methamphetamine 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 24502215-0 2014 Effect of 2-amino-1-methyl-6-phenylimidazo [4, 5-b] pyridine on oxidative stress and gene expression of c-fos, c-jun, p16 and Rb in rat colons and protective role of seabuckthorn seed oil. 2-amino-1-methyl-6-phenylimidazo(4,5-b)pyridine 10-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 24401792-9 2014 The number of c-Fos-IR cells in the VcI/II at every level (0-3.6 mm caudal to the obex) after formalin injection was significantly decreased (p < 0.01) with preadministration of morphine (3 mg/kg) or buprenorphine (100 microg/kg) in the control rats. Formaldehyde 94-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 24269295-7 2014 Expression of the c-fos mRNA in the arcuate nucleus, ventromedial hypothalamic nucleus (VMH) and rostral ventrolateral medulla (RVLM) in UNC1-injected rats showed significantly higher expression than in the PBS-injected rats. Lead 207-210 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 24401792-9 2014 The number of c-Fos-IR cells in the VcI/II at every level (0-3.6 mm caudal to the obex) after formalin injection was significantly decreased (p < 0.01) with preadministration of morphine (3 mg/kg) or buprenorphine (100 microg/kg) in the control rats. Morphine 181-189 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 24401792-9 2014 The number of c-Fos-IR cells in the VcI/II at every level (0-3.6 mm caudal to the obex) after formalin injection was significantly decreased (p < 0.01) with preadministration of morphine (3 mg/kg) or buprenorphine (100 microg/kg) in the control rats. Buprenorphine 203-216 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 24118285-8 2013 PPT also increased Fos expression within the PVN of EPM-exposed females; however, both vehicle- and PPT-treated primiparous females had reduced Fos expression compared to nulliparous females. 4,4',4''-(4-propyl-((1)H)-pyrazole-1,3,5-triyl) tris-phenol 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 24118285-8 2013 PPT also increased Fos expression within the PVN of EPM-exposed females; however, both vehicle- and PPT-treated primiparous females had reduced Fos expression compared to nulliparous females. 4,4',4''-(4-propyl-((1)H)-pyrazole-1,3,5-triyl) tris-phenol 100-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-147 24118285-9 2013 In the amygdala, PPT increased Fos immunoreactivity in the central but not the medial or basolateral amygdala, although these effects were only observed in home cage females. 4,4',4''-(4-propyl-((1)H)-pyrazole-1,3,5-triyl) tris-phenol 17-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 24118285-10 2013 Additionally, both vehicle- and PPT-treated home cage, primiparous females had increased Fos in the central nucleus of the amygdala compared to nulliparous controls. 4,4',4''-(4-propyl-((1)H)-pyrazole-1,3,5-triyl) tris-phenol 32-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 24132574-9 2013 At the end of the experiment, H2 excretion and the portal H2 concentration were significantly greater in the FOS group than in the control group. Hydrogen 30-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-112 24132574-9 2013 At the end of the experiment, H2 excretion and the portal H2 concentration were significantly greater in the FOS group than in the control group. Hydrogen 58-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-112 24132574-10 2013 In the FOS group, the arterial H2 concentration was no more than 1.5% of the portal H2 concentration (P = 0.03). Hydrogen 31-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 7-10 24132574-11 2013 The H2 concentration in abdominal cavity tissues, especially adipose tissue, in the FOS group was 5.6- to 43-fold of that in the control group (P < 0.05). Hydrogen 4-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 24132574-12 2013 The H2 content in the abdominal cavity in the FOS group was 11-fold of that in the control group (P < 0.05). Hydrogen 4-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 24132574-17 2013 Reduced cytokine expression by FOS feeding might be dependent on increased colonic H2. Hydrogen 83-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 23535348-6 2013 Fos protein immunoreactivity revealed that contextual fear promoted wide activation of the mPFC, which was significantly reduced after intra-DH infusions of muscimol. Muscimol 157-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 23585120-9 2013 c-Fos mRNA levels induced by thrombin were reduced by PD98059, SP600125, and Go6976, but not SB202190. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 54-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 24225225-1 2013 BACKGROUND: Hypothalamic neuropeptide Y (NPY) and two immediate early genes, c-fos and c-jun, have been found to be involved in regulating the appetite-suppressing effect of amphetamine (AMPH). Amphetamine 174-185 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 24225225-1 2013 BACKGROUND: Hypothalamic neuropeptide Y (NPY) and two immediate early genes, c-fos and c-jun, have been found to be involved in regulating the appetite-suppressing effect of amphetamine (AMPH). Amphetamine 187-191 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 24011250-4 2013 Using immunohistochemistry on the brain of male rats, we show here that portal glucose increases c-Fos expression in the brainstem, in the hypothalamus (in particular in neurons expressing pro-opiomelanocortin) and also in olfactory and other limbic and cortical areas, including those functionally implicated in reward (Experiment 1). Glucose 79-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 24011250-5 2013 In similar postabsorptive conditions, a high-protein diet induced similar effects in the hypothalamus and the granular cells of the main olfactory bulb, whereas the high-fat/high-sucrose diet actually reduced the basal expression of c-Fos in cortical layers. Sucrose 179-186 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 233-238 23895375-0 2014 Detection of molecular alterations in methamphetamine-activated Fos-expressing neurons from a single rat dorsal striatum using fluorescence-activated cell sorting (FACS). Methamphetamine 38-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 23895375-5 2014 Fos and NeuN (a neuronal marker) immunohistochemistry indicate that 5-6% of dorsal striatum neurons were activated 90 min after acute methamphetamine injections (5 mg/kg, i.p.) Methamphetamine 134-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 23895375-8 2014 Methamphetamine induced 3-20-fold increases of immediate early genes arc, homer-2, c-fos, fosB, and its isoforms (DeltafosB and a novel isoform DeltafosB-2) in Fos-positive but not Fos-negative neurons. Methamphetamine 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 23895375-8 2014 Methamphetamine induced 3-20-fold increases of immediate early genes arc, homer-2, c-fos, fosB, and its isoforms (DeltafosB and a novel isoform DeltafosB-2) in Fos-positive but not Fos-negative neurons. Methamphetamine 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-163 23895375-8 2014 Methamphetamine induced 3-20-fold increases of immediate early genes arc, homer-2, c-fos, fosB, and its isoforms (DeltafosB and a novel isoform DeltafosB-2) in Fos-positive but not Fos-negative neurons. Methamphetamine 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 181-184 23895375-13 2014 We used FACS along with targeted PCR pre-amplification to assess acute methamphetamine-induced gene expression from as few as 5 Fos-expressing neurons from a single rat dorsal striatum. Methamphetamine 71-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-131 23895375-14 2014 Methamphetamine induced 3-20-fold increases of immediate early genes (IEGs) in Fos-positive but not Fos-negative neurons. Methamphetamine 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 23810829-6 2013 Intraplantar formalin caused robust neurokinin 1 receptor (NK1r) internalization (indicating SP release) and c-Fos expression in the ipsilateral dorsal horn, which were blocked by IT SNX-482. Formaldehyde 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 23857113-4 2013 OBJECTIVE: The aim of the present study was to examine brain neuronal activation in female rats treated with flibanserin, using single immunocytochemical labeling of Fos protein, a marker of neuronal activation, and co-localization of Fos and catecholaminergic marker. flibanserin 109-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-169 23857113-4 2013 OBJECTIVE: The aim of the present study was to examine brain neuronal activation in female rats treated with flibanserin, using single immunocytochemical labeling of Fos protein, a marker of neuronal activation, and co-localization of Fos and catecholaminergic marker. flibanserin 109-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 235-238 23857113-7 2013 RESULTS: Acute flibanserin increased levels of Fos immunoreactivity in the nucleus accumbens, arcuate hypothalamic nucleus, locus coeruleus, lateral paragigantocellular nucleus, and nucleus of the solitary tract. flibanserin 15-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 23857113-8 2013 Chronic 22-day treatment with flibanserin increased Fos expression in the medial preoptic area and arcuate nucleus of the hypothalamus, ventral tegmental area, locus coeruleus, and lateral paragigantocellular nucleus. flibanserin 30-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 23766142-0 2013 Fos expression in response to dopamine D3-preferring phenylpiperazine drugs given with and without cocaine. Dopamine 30-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 23766142-0 2013 Fos expression in response to dopamine D3-preferring phenylpiperazine drugs given with and without cocaine. phenylpiperazine 53-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 23766142-0 2013 Fos expression in response to dopamine D3-preferring phenylpiperazine drugs given with and without cocaine. Cocaine 99-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 24383084-0 2013 Neuronal Fos-like immunoreactivity associated with dexamethasone-induced hypertension in rats and effects of glucagon-like peptide-2. Dexamethasone 52-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 24383084-9 2013 KEY FINDINGS: Fos-immunoreactivity (Fos-IR) in the dorsomedial hypothalamic nucleus (DMH) was higher in dexamethasone-treated rats than in saline-treated rats. Dexamethasone 104-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 24383084-9 2013 KEY FINDINGS: Fos-immunoreactivity (Fos-IR) in the dorsomedial hypothalamic nucleus (DMH) was higher in dexamethasone-treated rats than in saline-treated rats. Dexamethasone 104-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-39 24383084-12 2013 After the peripheral administration of GLP-2, Fos-IR in the caudal ventrolateral medulla (CVLM) increased in dexamethasone-treated rats. Dexamethasone 109-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 24383084-13 2013 SIGNIFICANCE: Chronic dexamethasone treatment induced Fos-IR in the DMH. Dexamethasone 22-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 24049118-9 2013 Catecholaminergic cells colabeled with Fos immunoreactivity in the CVLM were observed following 12% O2, and further increases in hypoxia severity caused markedly more activation. Oxygen 100-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 24064338-2 2013 Our previous work has shown that peripherally administered OEA activates c-fos transcription in the nucleus of the solitary tract (NST) and in the paraventricular nucleus (PVN), where it enhances oxytocin (OXY) expression. oleoylethanolamide 59-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 24064338-7 2013 We found that the DSAP lesion completely prevented OEA"s effects on food intake, on Fos and OXY expression in the PVN, and on OXY immunoreactivity of the posterior pituitary gland; all effects were maintained in sham-operated rats. dsap 18-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 23585120-9 2013 c-Fos mRNA levels induced by thrombin were reduced by PD98059, SP600125, and Go6976, but not SB202190. pyrazolanthrone 63-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 23585120-9 2013 c-Fos mRNA levels induced by thrombin were reduced by PD98059, SP600125, and Go6976, but not SB202190. Go 6976 77-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 23585120-10 2013 Thrombin stimulated in vivo binding of c-Fos to the MMP-9 promoter, which was reduced by pretreatment with SP600125 or PD98059, but not SB202190. pyrazolanthrone 107-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 23585120-10 2013 Thrombin stimulated in vivo binding of c-Fos to the MMP-9 promoter, which was reduced by pretreatment with SP600125 or PD98059, but not SB202190. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 119-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 23368947-10 2013 Surprisingly, TCN enhanced the expression of c-fos and amphiregulin mRNA. Triclosan 14-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 24060804-3 2013 We hypothesize that as mediators of the expression of MMP-9 the transcription factors like nuclear factor kappa B (NF-kappaB), c-fos and retinoic acid receptors-alpha (RAR-alpha) with binding sites to the MMP-9 promoter are overexpressed in the spontaneously hypertensive rat (SHR) in a process that is regulated by oxygen free radicals. oxygen free radicals 316-336 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-132 24060804-9 2013 These results suggest that elevated NF-kappaB, c-fos and RAR-alpha expressions and MMP-9 activity in the SHR are superoxide-dependent. Superoxides 113-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 23959001-2 2013 This effect, which requires intact vagal fibers and intestinal PPAR-alpha receptors, is coupled to the increase of c-fos and oxytocin mRNA expression in neurons of the paraventricular nucleus (PVN) and is prevented by the intracerebroventricular administration of a selective oxytocin antagonist, thus suggesting a necessary role of oxytocinergic neurotransmission in the pro-satiety effect of OEA. oleoylethanolamide 394-397 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 24109472-4 2013 At the cellular level, DA induces the expression of learning-related genes via the transcription factor c-Fos. Dopamine 23-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 24039778-2 2013 We hypothesized that quercetin inhibits cardiac hypertrophy by blocking AP-1 (c-fos, c-jun) and activating PPAR-gamma signaling pathways. Quercetin 21-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 23922220-0 2013 Imipramine-induced c-Fos expression in the medial prefrontal cortex is decreased in the ACTH-treated rats. Imipramine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 23922220-4 2013 In further study, imipramine produced significant increases in the c-Fos expression in the medial prefrontal cortex (mPFC), the dentate gyrus of the hippocampus (DGH), and the central nucleus of the amygdala (CeA), in rats repeatedly treated with saline. Imipramine 18-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 23922220-4 2013 In further study, imipramine produced significant increases in the c-Fos expression in the medial prefrontal cortex (mPFC), the dentate gyrus of the hippocampus (DGH), and the central nucleus of the amygdala (CeA), in rats repeatedly treated with saline. Sodium Chloride 247-253 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 23922220-5 2013 The imipramine-increased c-Fos immunoreactivity was suppressed in the mPFC of rats repeatedly treated with ACTH. Imipramine 4-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 24073217-0 2013 Body sodium overload modulates the firing rate and fos immunoreactivity of serotonergic cells of dorsal raphe nucleus. Sodium 5-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 24073217-7 2013 Both Fos-OT immunoreactive and plasma OT concentration increased after s.c. hypertonic sodium infusion. Sodium 87-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 5-8 23806722-13 2013 bLRs" increase in positive affect after chronic EC was coupled with significant positive correlations between corticosterone levels and c-fos mRNA in the accumbens. Corticosterone 110-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 23806722-14 2013 Conversely, a decline in the rate of positive calls in bHRs after chronic EC was associated with a negative correlation between corticosterone and accumbens c-fos mRNA. Corticosterone 128-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-162 24056223-6 2013 Pre-administration of the QX cocktail significantly reduced to sham levels Fos-LI examined 2 hrs after tooth extraction; reduced Fos-LI was also observed with the conventional local anesthetic lidocaine. Lidocaine 193-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-132 23681297-3 2013 The 5-HT2C antagonist SB243,213 (1 mg/kg), which enhanced Fos per se in the striatum and the subthalamic nucleus (STN) only, was used to study the implication of 5-HT2C receptors. sb243 22-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 23681297-4 2013 The agonists Ro 60-0175 (3 mg/kg) and m-CPP (1 mg/kg) and the inverse agonist SB206,553 (10 mg/kg) enhanced Fos expression in the STN and faintly in the entopeduncular nucleus (EPN, the internal globus pallidus in primate). Ro 60-0175 13-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 23681297-4 2013 The agonists Ro 60-0175 (3 mg/kg) and m-CPP (1 mg/kg) and the inverse agonist SB206,553 (10 mg/kg) enhanced Fos expression in the STN and faintly in the entopeduncular nucleus (EPN, the internal globus pallidus in primate). sb206 78-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 23681297-7 2013 Their Fos effect in the STN was reduced significantly by SB243,213 only in the case of m-CPP. sb243 57-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 6-9 23900858-8 2013 As expected, c-Fos-immunopositive cells were also found in these regions, suggesting that increased FDG signals induced by intraoral citrate injection are likely to reflect neural activity in these regions. Citric Acid 133-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 24250203-0 2013 Expression of cFos and brain-derived neurotrophic factor in cortex and hippocampus of ethanol-withdrawn male and female rats. Ethanol 86-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-18 23726845-7 2013 METH self-administration caused increases in mRNA expression of the transcription factors, c-fos and fosb, the neurotrophic factor, Bdnf, and the synaptic protein, synaptophysin (Syp) in the dorsal striatum. Methamphetamine 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-105 23726845-9 2013 Importantly, ChIP-PCR showed that METH self-administration caused enrichment of phosphorylated CREB (pCREB), but not of histone H3 trimethylated at lysine 4 (H3K4me3), on promoters of c-fos, fosb, Bdnf and Syp at 2h after cessation of drug intake. Methamphetamine 34-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 184-189 23774134-10 2013 Fluoxetine (10 mg/kg) stimulated greater increases in c-Fos expression across the extended amygdala in adults than in adolescents, and 8-OH DPAT (0.5 mg/kg) produced greater increases in c-Fos in the lateral orbital cortex and central nucleus of the amygdala in adults. Fluoxetine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 23774134-10 2013 Fluoxetine (10 mg/kg) stimulated greater increases in c-Fos expression across the extended amygdala in adults than in adolescents, and 8-OH DPAT (0.5 mg/kg) produced greater increases in c-Fos in the lateral orbital cortex and central nucleus of the amygdala in adults. 8-Hydroxy-2-(di-n-propylamino)tetralin 135-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 187-192 24039778-7 2013 Conversely, in vivo, AP-1 (c-fos, s-jun) activation was suppressed in the quercetin-treated group, as was the downstream hypertrophy gene, including mRNA levels of ANP and BNP (P<0.05, vs. SHRs). Quercetin 74-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 23701881-7 2013 Fasting-induced Fos expression accompanying endogenous brainstem TRH action decreased by 66% and 91%, respectively, in the nucleus tractus solitarius (NTS) and the dorsal motor nucleus of the vagus (DMV) in GK rats, compared to Wistar rats. (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 199-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 23764459-5 2013 Our results indicate that exposure to FLX influenced restraint stress-induced Fos expression in the amygdala in a gender and age-specific manner. Fluoxetine 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 23938388-7 2013 Oral capsaicin exposure significantly increased c-Fos expression not only in the nucleus tractus of solitarius but also in the paraventricular nucleus. Capsaicin 5-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 23707289-9 2013 Yohimbine reversibly unmasked contralateral hindlimb allodynia and hyperalgesia of all modalities and increased dorsal horn c-fos expression to an innocuous brush stimulus. Yohimbine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 24308186-0 2013 [Effect of electroacupuncture intervention at different time-points in a day on expression of c-fos and neuronal nitric oxide synthase in medial prefrontal cortex in ketamine addiction rats]. Ketamine 166-174 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 24308186-1 2013 UNLABELLED: To observe the effects of electroacupuncture(EA) stimulation of "Zusanli"(ST 36) and "Sanyinjiao"(SP 6) at different time-points of a day on the expression of c-fos and neuronal nitric oxide synthase (nNOS) in the medial prefrontal cortex (mPFC) in rats with ketamine addiction. Ketamine 271-279 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 171-176 24308186-8 2013 Compared with the normal control group and saline group, the expression levels of c-fos and nNOS in the mPFC in the model group were significantly upregulated (P < 0.05). Sodium Chloride 43-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 24093120-8 2013 RESULTS: IA acetaminophen decreased both the severity and distribution of c-Fos expression. Acetaminophen 12-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 24093120-9 2013 IP acetaminophen decreased only the distribution of c-Fos expression. Acetaminophen 3-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 23447367-6 2013 c-Fos expression in NAcc, but not in CPu, was associated with these alterations in cocaine sensitization. nacc 20-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 23911956-4 2013 injection of either capsaicin or AITC (0.03-10 mg/kg) induced short-term (up to 20 min) and dose-dependent abdominal nociception, and also produced c-fos expression in spinal afferents of the dorsal horn. Capsaicin 20-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 23911956-4 2013 injection of either capsaicin or AITC (0.03-10 mg/kg) induced short-term (up to 20 min) and dose-dependent abdominal nociception, and also produced c-fos expression in spinal afferents of the dorsal horn. 2,3,4-tri-O-acetylarabinopyranosyl isothiocyanate 33-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 23806600-0 2013 Effect of blonanserin on methamphetamine-induced disruption of latent inhibition and c-Fos expression in rats. blonanserin 10-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 23806600-0 2013 Effect of blonanserin on methamphetamine-induced disruption of latent inhibition and c-Fos expression in rats. Methamphetamine 25-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 23806600-8 2013 increased c-Fos expression in the shell area but not in the core area of the nucleus accumbens while methamphetamine (3.0mg/kg, i.p.) Methamphetamine 101-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-15 23806600-10 2013 Blonanserin also increased the number of c-Fos expressions in the central amygdala nucleus but not in the basolateral amygdala nucleus or the prefrontal cortex. blonanserin 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 23714385-8 2013 Bioinformatics analyses indicated alterations in a) transcription factors that reduced excitation-coupled transcription and promoted excitotoxic neuronal damage involving clusters of genes associated with Nfkbia, Fos, and Srebf1, b) genes that reduced cholesterol and fatty acid synthesis, and increased protein degradation, and c) genes involved in cell-to-cell interactions and regulation of the actin cytoskeleton. Cholesterol 252-263 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-216 23714385-8 2013 Bioinformatics analyses indicated alterations in a) transcription factors that reduced excitation-coupled transcription and promoted excitotoxic neuronal damage involving clusters of genes associated with Nfkbia, Fos, and Srebf1, b) genes that reduced cholesterol and fatty acid synthesis, and increased protein degradation, and c) genes involved in cell-to-cell interactions and regulation of the actin cytoskeleton. Fatty Acids 268-278 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-216 23708866-10 2013 All agents reduced c-Fos expression (control: 34 +- 4, fentanyl: 8 +- 2, isoflurane: 12 +- 3, nitrous oxide: 11 +- 2, isoflurane + nitrous oxide: 12 +- 1, pentobarbital: 11 +- 2, propofol: 13 +- 3; P < 0.05; n = 3). Nitrous Oxide 131-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 23692698-8 2013 MCS decreased Fos immunoreactivity (Fos-IR) in the superficial layers of the dorsal horn of the spinal cord for all evaluated groups and increased Fos-IR in the PAG, although no changes were observed in the PAG for the tail group. mcs 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 23692698-8 2013 MCS decreased Fos immunoreactivity (Fos-IR) in the superficial layers of the dorsal horn of the spinal cord for all evaluated groups and increased Fos-IR in the PAG, although no changes were observed in the PAG for the tail group. mcs 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-39 23692698-8 2013 MCS decreased Fos immunoreactivity (Fos-IR) in the superficial layers of the dorsal horn of the spinal cord for all evaluated groups and increased Fos-IR in the PAG, although no changes were observed in the PAG for the tail group. mcs 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-39 23640815-5 2013 We hypothesized that MCs would exhibit c-fos expression even if rats were examined randomly, under normal housing conditions. mcs 21-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 23640815-8 2013 C-fos-ir hilar cells co-expressed GluR2/3, suggesting that they were MCs. mcs 69-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 23640815-10 2013 However, c-fos-ir in dorsal MCs was reduced under these circumstances, suggesting that ventral and dorsal MCs are functionally distinct. mcs 28-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 23640815-11 2013 Interestingly, there was an inverse relationship between MC and GC layer c-fos expression, with little c-fos expression in the GC layer in ventral sections where MC expression was strong, and the opposite in dorsal hippocampus. Methylcholanthrene 57-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 23825033-3 2013 E advanced AICAR-induced increases in A2 phospho-AMPK (pAMPK) expression and in blood glucose levels and was required for augmentation of Fos, estrogen receptor-alpha (ERalpha), monocarboxylate transporter-2, and glucose transporter-3 protein in A2 neurons and enhancement of corticosterone secretion by this treatment paradigm. acadesine 11-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-141 23770308-6 2013 Furthermore, KA-induced c-Fos expression and extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation in the hippocampus were also reduced by bupropion pretreatment. Bupropion 152-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 23716620-8 2013 Remarkably, the number of c-Fos(+) neurons in ventral motor thalamic nuclei was higher in PRS rats than in unstressed controls, both under basal conditions and in response to single or repeated injections with haloperidol. Haloperidol 210-221 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 23946692-0 2013 Changes in c-Fos Expression in the Forced Swimming Test: Common and Distinct Modulation in Rat Brain by Desipramine and Citalopram. Desipramine 104-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 23946692-0 2013 Changes in c-Fos Expression in the Forced Swimming Test: Common and Distinct Modulation in Rat Brain by Desipramine and Citalopram. Citalopram 120-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 23946692-4 2013 Desipramine treatment alone in the absence of pretest swim was without effect, whereas citalopram treatment alone significantly increased the number of c-Fos-like immunoreactive cells in the central nucleus of the amygdala and bed nucleus of the stria terminalis, where this pattern of increase appears to be maintained after the pretest swim. Citalopram 87-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-157 23946692-5 2013 Both desipramine and citalopram treatment after the pretest swim significantly increased the number of c-Fos-like immunoreactive cells in the ventral lateral septum and ventrolateral periaqueductal gray before the test swim. Desipramine 5-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 23946692-5 2013 Both desipramine and citalopram treatment after the pretest swim significantly increased the number of c-Fos-like immunoreactive cells in the ventral lateral septum and ventrolateral periaqueductal gray before the test swim. Citalopram 21-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 23946692-6 2013 These results suggest that citalopram may affect c-Fos expression in the central nucleus of the amygdala and bed nucleus of the stria terminalis distinctively and raise the possibility that upregulation of c-Fos in the ventral lateral septum and ventrolateral periaqueductal gray before the test swim may be one of the probable common mechanisms underlying antidepressant effect in the FST. Citalopram 27-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 23946692-6 2013 These results suggest that citalopram may affect c-Fos expression in the central nucleus of the amygdala and bed nucleus of the stria terminalis distinctively and raise the possibility that upregulation of c-Fos in the ventral lateral septum and ventrolateral periaqueductal gray before the test swim may be one of the probable common mechanisms underlying antidepressant effect in the FST. Citalopram 27-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 206-211 23922700-16 2013 The anti-angiogenesis effect afforded by celecoxib may attribute to its modulation on VEGF/VEGFR-2 through the down-regulation of integrated signal pathways involving PGE2- HIF-1alpha- VEGF and p-ERK- c-fos- VEGFR-2. Celecoxib 41-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 201-206 23500454-9 2013 Treatment with the insulin-sensitizing drug metformin attenuated estrogen-dependent proliferative expression of c-myc and c-fos in the obese rat endometrium compared to untreated controls and was accompanied by inhibition of phosphorylation of the insulin and IGF1 receptors (IRbeta/IGF1R) and ERK1/2. Metformin 44-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 23517225-6 2013 After injury, chondroitin sulfate proteoglycan digestion produced the expected increase in growth-associated protein 43-positive axons and perikarya, of which a significantly greater number were double labeled for c-Fos after intervention with chondroitinase, compared to vehicle. Chondroitin Sulfates 14-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 214-219 23653461-4 2013 Rats were rendered hypernatremic by acute administration of 2.0 M NaCl and had increased plasma sodium concentration, plasma osmolality, and Fos induction in OT-containing neurons relative to 0.15 M NaCl-treated controls. Sodium Chloride 66-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 23380305-0 2013 Selective brain region activation by histamine H3 receptor antagonist/inverse agonist ABT-239 enhances acetylcholine and histamine release and increases c-Fos expression. 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid 86-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-158 23380305-0 2013 Selective brain region activation by histamine H3 receptor antagonist/inverse agonist ABT-239 enhances acetylcholine and histamine release and increases c-Fos expression. Histamine 37-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-158 23380305-8 2013 Systemic as well as intra-TMN administration of ABT-239 increased c-Fos expression in the NBM, and cortex, but not in the striatum or NAcc. 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid 48-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 23603032-7 2013 Our results indicate that 2-AG increases REMS through CB1R activation, and increases c-Fos expression in MCH neurons. glyceryl 2-arachidonate 26-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 23490179-0 2013 Neurobehavioral effects, c-Fos/Jun expression and tissue distribution in rat offspring prenatally co-exposed to MeHg and PFOA: PFOA impairs Hg retention. perfluorooctanoic acid 121-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 23745109-0 2013 c-Fos immunoreactivity in prefrontal, basal ganglia and limbic areas of the rat brain after central and peripheral administration of ethanol and its metabolite acetaldehyde. Ethanol 133-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 23745109-0 2013 c-Fos immunoreactivity in prefrontal, basal ganglia and limbic areas of the rat brain after central and peripheral administration of ethanol and its metabolite acetaldehyde. Acetaldehyde 160-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 23745109-7 2013 IP administration of EtOH minimally induced c-Fos in some regions of the prefrontal cortex and basal ganglia, mainly at the low dose (0.5 g/kg), while IP acetaldehyde induced c-Fos in virtually all the structures studied at both doses. Ethanol 21-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 23745109-7 2013 IP administration of EtOH minimally induced c-Fos in some regions of the prefrontal cortex and basal ganglia, mainly at the low dose (0.5 g/kg), while IP acetaldehyde induced c-Fos in virtually all the structures studied at both doses. Acetaldehyde 154-166 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 175-180 23745109-8 2013 Acetaldehyde administered centrally increased c-Fos in all areas studied, a pattern that was very similar to EtOH. Acetaldehyde 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 23745109-9 2013 Thus, IP administered acetaldehyde was more efficacious than EtOH at inducing c-Fos expression. Acetaldehyde 22-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 23745109-9 2013 Thus, IP administered acetaldehyde was more efficacious than EtOH at inducing c-Fos expression. Ethanol 61-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 23745109-10 2013 However, the general pattern of c-Fos induction promoted by ICV EtOH and acetaldehyde was similar. Ethanol 64-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 23745109-10 2013 However, the general pattern of c-Fos induction promoted by ICV EtOH and acetaldehyde was similar. Acetaldehyde 73-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 27335881-2 2013 Following application of the pain producing substance (PPS) infusion, the number of Fos-labeled cells increased significantly in the subnucleus caudalis (Sp5C) compared with other nuclei in the TSNC. pps 55-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 23818940-8 2013 Administration of losartan and more strongly moxonidine modulated most effects and particularly inhibited Fos-activity in locus coeruleus neurons. Losartan 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 23818940-8 2013 Administration of losartan and more strongly moxonidine modulated most effects and particularly inhibited Fos-activity in locus coeruleus neurons. moxonidine 45-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 23558307-6 2013 RF rats exhibited a food-entrained rhythm of c-Fos in ORX cells in the DMH, LH and PeF with highest levels at the time of meal delivery and after food ingestion. Dimenhydrinate 71-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 23551217-3 2013 Here, we describe the effect of DCS administration, alone or in conjunction with extinction training, on neuronal activity (c-fos) and neuronal plasticity [phospho-extracellular signal-regulated kinase (pERK)] markers using immunohistochemistry. Cycloserine 32-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 23551217-4 2013 We found that intraperitoneal administration of DCS in untrained young rats (24-28 days old) increased c-fos- and pERK-stained neurons in both the prelimbic and infralimbic division of the medial prefrontal cortex (mPFC) and reduced pERK levels in the lateral nucleus of the central amygdala. Cycloserine 48-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 22964037-6 2013 SKF38393, quinpirole and sulpiride induced no behavioural changes, but the D1-like receptor antagonist SCH23390 increased the freezing response of the rats and selectively reduced Fos protein expression in the anterior cingulate cortex and rostral NAcSh. SCH 23390 103-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 180-183 23499563-6 2013 The present study was designed to evaluate the acute effects of clozapine (atypical), chlorpromazine (typical) and fluphenazine (typical) on c-Fos expression (a marker of neuronal response) in these appetite-related centers of the rat brain. Clozapine 64-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 23499563-6 2013 The present study was designed to evaluate the acute effects of clozapine (atypical), chlorpromazine (typical) and fluphenazine (typical) on c-Fos expression (a marker of neuronal response) in these appetite-related centers of the rat brain. Chlorpromazine 86-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 23499563-6 2013 The present study was designed to evaluate the acute effects of clozapine (atypical), chlorpromazine (typical) and fluphenazine (typical) on c-Fos expression (a marker of neuronal response) in these appetite-related centers of the rat brain. Fluphenazine 115-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 23499563-8 2013 Acute treatment with clozapine, chlorpromazine and fluphenazine differentially influenced c-Fos expression in these brain structures. Clozapine 21-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 23499563-8 2013 Acute treatment with clozapine, chlorpromazine and fluphenazine differentially influenced c-Fos expression in these brain structures. Chlorpromazine 32-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 23499563-8 2013 Acute treatment with clozapine, chlorpromazine and fluphenazine differentially influenced c-Fos expression in these brain structures. Fluphenazine 51-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 23593280-0 2013 Lesions of the fasciculus retroflexus alter footshock-induced cFos expression in the mesopontine rostromedial tegmental area of rats. mesopontine 85-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-66 22927669-10 2013 HAL, but not OLZ, also enhanced AMPH-induced psychomotor activation and c-fos mRNA expression in the caudate-putamen. Haloperidol 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 23616555-5 2013 c-Fos experiments further showed that cocaine-activated LHb neurons preferentially projected to and activated neurons in the rostromedial tegmental nucleus (RMTg), a recently identified target of LHb axons that is activated by negative motivational stimuli and inhibits dopamine neurons. 5-[[[(2~{r},3~{s},4~{r},5~{r})-5-(6-Aminopurin-9-Yl)-3,4-Bis(Oxidanyl)oxolan-2-Yl]methylamino]methyl]-4-Azanyl-1-(Methoxymethyl)pyrimidin-2-One 56-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 23616555-5 2013 c-Fos experiments further showed that cocaine-activated LHb neurons preferentially projected to and activated neurons in the rostromedial tegmental nucleus (RMTg), a recently identified target of LHb axons that is activated by negative motivational stimuli and inhibits dopamine neurons. rmtg 157-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 23616555-5 2013 c-Fos experiments further showed that cocaine-activated LHb neurons preferentially projected to and activated neurons in the rostromedial tegmental nucleus (RMTg), a recently identified target of LHb axons that is activated by negative motivational stimuli and inhibits dopamine neurons. 5-[[[(2~{r},3~{s},4~{r},5~{r})-5-(6-Aminopurin-9-Yl)-3,4-Bis(Oxidanyl)oxolan-2-Yl]methylamino]methyl]-4-Azanyl-1-(Methoxymethyl)pyrimidin-2-One 196-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 23499799-9 2013 There was a marked 100-fold increase in cFOS mRNA expression and 100-fold decrease in NK2 mRNA expression in the whole blood of capsaicin-treated rats. Capsaicin 128-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-44 23593280-9 2013 These data confirm the involvement of the lateral habenula in modulating the activity of rostromedial tegmental area neurons in response to mild aversive stimuli and suggest that dopamine input may contribute to footshock- induced activation of cFos expression in the lateral habenula. Dopamine 179-187 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 245-249 23328445-4 2013 The results demonstrate that one iTBS block may have an after-effect of at least two different phases, an early phase with increased neuronal activity (c-Fos, zif268) but also the likelihood of increased GABA-release (GAD65), followed by a late phase (>40min) of reduced neuronal activity in excitatory and inhibitory systems which may indicate a state of reduced excitability. itbs 33-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 152-157 23328070-9 2013 In addition, c-jun and c-fos expression in the hippocampus of 40-day-old rats was decreased in marginal iodine-deficient rats, compared with control. Iodine 104-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 23270755-3 2013 A2NTX also reduced the increase in c-fos immunoreactivity in L4-L5 spinal segments induced by carrageenan, suggesting that A2NTX inhibits the activation of spinal nociceptive afferent fibers that project to the CNS. a2ntx 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 23270755-3 2013 A2NTX also reduced the increase in c-fos immunoreactivity in L4-L5 spinal segments induced by carrageenan, suggesting that A2NTX inhibits the activation of spinal nociceptive afferent fibers that project to the CNS. Carrageenan 94-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 23270755-3 2013 A2NTX also reduced the increase in c-fos immunoreactivity in L4-L5 spinal segments induced by carrageenan, suggesting that A2NTX inhibits the activation of spinal nociceptive afferent fibers that project to the CNS. a2ntx 123-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 23239012-4 2013 The double labeling immunohistochemistry combined with c-Fos detection as a marker of neuronal activation revealed significantly higher numbers of c-Fos-positive vasopressinergic neurons both in the supraoptic and paraventricular nuclei and tyrosine hydroxylase containing medullary A1 neurons, of L-cysteine-injected rats than those injected with D-cysteine as iso-osmotic control. Cysteine 298-308 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-152 23239012-4 2013 The double labeling immunohistochemistry combined with c-Fos detection as a marker of neuronal activation revealed significantly higher numbers of c-Fos-positive vasopressinergic neurons both in the supraoptic and paraventricular nuclei and tyrosine hydroxylase containing medullary A1 neurons, of L-cysteine-injected rats than those injected with D-cysteine as iso-osmotic control. D-cysteine 348-358 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-152 23054911-10 2013 The weight percentage fraction of infarction size was increased after c-fos was increased via the administration of isoproterenol. Isoproterenol 116-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 23054911-11 2013 c-Fos protein expression and the infarct size in rats treated with metoprolol were also decreased compared with the control normal saline treatment group. Metoprolol 67-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 23054911-13 2013 Metoprolol can inhibit the expression of c-fos and has a positive therapeutic effect on rats after AMI; the involvement effect of metoprolol on myocardial infarction might be correlated with its effect on the inhibition of c-fos. Metoprolol 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 23054911-13 2013 Metoprolol can inhibit the expression of c-fos and has a positive therapeutic effect on rats after AMI; the involvement effect of metoprolol on myocardial infarction might be correlated with its effect on the inhibition of c-fos. Metoprolol 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 223-228 23054911-13 2013 Metoprolol can inhibit the expression of c-fos and has a positive therapeutic effect on rats after AMI; the involvement effect of metoprolol on myocardial infarction might be correlated with its effect on the inhibition of c-fos. Metoprolol 130-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 23054911-13 2013 Metoprolol can inhibit the expression of c-fos and has a positive therapeutic effect on rats after AMI; the involvement effect of metoprolol on myocardial infarction might be correlated with its effect on the inhibition of c-fos. Metoprolol 130-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 223-228 22893527-0 2013 Dynamic variations of c-Fos expression in the spinal cord exposed to intramuscular hypertonic saline-induced muscle nociception. Sodium Chloride 94-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 22893527-8 2013 5.8% saline injection, c-Fos expression in dorsal horn of spinal L4-6 segments was significantly enhanced bilaterally (p < 0.05 and p < 0.001). Sodium Chloride 5-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 22893527-9 2013 These 5.8% saline-induced bilateral spinal Fos expression occurred rapidly 0.5 h following the i.m. Sodium Chloride 11-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 22893527-12 2013 However, the 5.8% saline-induced increases in Fos expression in intact and spinalized rats differed significantly. Sodium Chloride 18-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 22893527-14 2013 Fentanyl (20 mug/kg, intraperitoneal) completely attenuated the 5.8% saline intramuscularly induced increases in c-Fos expression in laminae III-VI (p < 0.001), but not laminae I-II. Fentanyl 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 22893527-14 2013 Fentanyl (20 mug/kg, intraperitoneal) completely attenuated the 5.8% saline intramuscularly induced increases in c-Fos expression in laminae III-VI (p < 0.001), but not laminae I-II. Sodium Chloride 69-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 23333670-0 2013 c-Fos activity mapping reveals differential effects of noradrenaline and serotonin depletion on the regulation of ocular dominance plasticity in rats. Norepinephrine 55-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 23333670-0 2013 c-Fos activity mapping reveals differential effects of noradrenaline and serotonin depletion on the regulation of ocular dominance plasticity in rats. Serotonin 73-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 23333670-3 2013 Applying this new method, here we studied the unique modification of the degree of c-Fos expression induced in the visual cortex, in that endogenous noradrenaline (NA) and serotonin (5HT) in the cortex were significantly reduced, respectively by specific pharmacological agents. Norepinephrine 149-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 23333670-3 2013 Applying this new method, here we studied the unique modification of the degree of c-Fos expression induced in the visual cortex, in that endogenous noradrenaline (NA) and serotonin (5HT) in the cortex were significantly reduced, respectively by specific pharmacological agents. Serotonin 172-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 23333670-3 2013 Applying this new method, here we studied the unique modification of the degree of c-Fos expression induced in the visual cortex, in that endogenous noradrenaline (NA) and serotonin (5HT) in the cortex were significantly reduced, respectively by specific pharmacological agents. Serotonin 183-186 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 23364524-8 2013 We found that fewer DBH cells expressed c-Fos in CIH- than in sham-treated rats in the medulla (significant in the A1 group). cih 49-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 23565937-6 2013 Here, using a female rat model, we show that the mPOA innervates the VTA in a region-specific manner, that lesions of the mPOA augment cocaine-induced Fos expression in the nucleus accumbens (NAc) and cocaine-induced conditioned place preference. Cocaine 135-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-154 23626473-0 2013 Ginsenosides Have a Suppressive Effect on c-Fos Expression in Brain and Reduce Cardiovascular Responses Increased by Noxious Stimulation to the Rat Tooth. Ginsenosides 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 23626473-7 2013 Ginsenosides reduced the number of c-Fos-IR increased by NS to tooth in the trigeminal Vc and thalamic VPMN and CLN. Ginsenosides 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 23626473-9 2013 Ginsenosides ameliorated arterial BP and HR raised by NS to tooth and reduced the number of Fos-IR neurons increased by NS to tooth in the NTS, RVLM, hypothalamic SON, and PVN. Ginsenosides 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 23287538-9 2013 This vulnerability to ethanol abuse was associated with a lower c-Fos immunoreactivity in the Nac and enduring alterations of the expression of Penk and Slc6a4, 2 neurotransmission-related genes that have been shown to play critical roles in the behavioral effects of ethanol and alcoholism. Ethanol 22-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 23438370-17 2013 Similarly, LiCl (76 mg/kg ip) or AngII (50 mug/kg sc) led to c-Fos expression only in CTR negative AP neurons. Lithium Chloride 11-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 23333602-0 2013 Study on the expression of c-Fos protein in the brain of rats after ingestion of food rich in lycopene. Lycopene 94-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 23375893-5 2013 Yet, upon first reencounter in a novel sodium-depletion state to promote mesocorticolimbic reactivity (reflected by elevated Fos activation in ventral tegmentum, nucleus accumbens, ventral pallidum, and the orbitofrontal prefrontal cortex), the learned cue was instantly transformed into an attractive and powerful motivational magnet. Sodium 39-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-128 23096770-5 2013 Finally, expression of the immediate early gene c-fos in Orx neurons was examined after self-administration, late extinction or cue-induced reinstatement of sucrose seeking. Sucrose 157-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 23295904-4 2013 In the present study we confirm that testosterone exposure during the neonatal period results in changes to a variety of typical aspects of the female reproductive system, including estrous cyclicity as shown by virginal smear, the positive feedback effects of estrogen alone or combined with progesterone, luteinizing hormone secretions, and estrogen and progesterone-induced Fos expression in gonadotropin-releasing hormone neurons. Testosterone 37-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 377-380 23098798-6 2013 Cocaine-induced c-Fos protein expression was augmented only in the ipsilateral nucleus accumbens core after mSTN lidocaine pretreatment, consistent with the expectation that inactivation of mSTN would disinhibit nucleus accumbens core, but not shell, activity. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 23183042-3 2013 The periaqueductal gray represents the final common pathway mediating defensive reactions to fear and we have reported previously that the dorsolateral PAG (dlPAG) exhibits a small but reliable increase in neural activity (as measured by c-fos protein immunoreactivity) when spontaneous recovery (SR) of a conditioned taste aversion (CTA) is reduced. phenylacetylglycine 152-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 238-243 23183042-11 2013 These data refute a cause-and-effect relationship between enhanced dPAG c-fos expression and a reduction in SR. dpag 67-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 22913654-6 2013 RESULTS: Data showed a reduced expression of NTG-induced Fos protein in the paraventricular nucleus (PVH), supraoptic nucleus (SON), and nucleus trigeminalis caudalis (SPVC) of male rats in comparison with female rats. Nitroglycerin 45-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 22913654-7 2013 Furthermore, in castrated female rats, NTG-induced neuronal activation was reduced in PVH, SON, central nucleus of the amygdala (AMI), nucleus tractus solitarius (NTS), area postrema (AP), and SPVC, while in castrated male rats Fos expression was reduced uniquely in the SPVC. Nitroglycerin 39-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 228-231 23098798-6 2013 Cocaine-induced c-Fos protein expression was augmented only in the ipsilateral nucleus accumbens core after mSTN lidocaine pretreatment, consistent with the expectation that inactivation of mSTN would disinhibit nucleus accumbens core, but not shell, activity. Lidocaine 113-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 23098799-0 2013 Effects of chronic treatment with corticosterone and imipramine on fos immunoreactivity and adult hippocampal neurogenesis. Corticosterone 34-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 23098799-0 2013 Effects of chronic treatment with corticosterone and imipramine on fos immunoreactivity and adult hippocampal neurogenesis. Imipramine 53-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 23098799-6 2013 Imipramine, on the other hand, increased fos-ir in the medial amygdala and decreased fos-ir in the anterior hypothalamus. Imipramine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 23098799-6 2013 Imipramine, on the other hand, increased fos-ir in the medial amygdala and decreased fos-ir in the anterior hypothalamus. Imipramine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 23072838-8 2013 Similarly, the adolescent-onset group failed to show significant neural activation in the prelimbic or infralimbic mPFC during the heroin-seeking test, whereas the adult-onset heroin self-administration group showed two to six times more Fos-ir(+) neurons than their saline counterparts in both mPFC subregions. Heroin 176-182 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 238-241 23266368-0 2013 Prenatal ethanol exposure increases ethanol intake and reduces c-Fos expression in infralimbic cortex of adolescent rats. Ethanol 9-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 23153991-0 2013 Differential regulation of CDK5 and c-Fos expression by morphine in the brain of Lewis and Fischer 344 rat strains. Morphine 56-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 23153991-1 2013 The aim of this study was to comparatively study cyclin-dependent kinase 5 (CDK5) and c-Fos regulation by morphine in the brains of Lewis and Fischer 344 (F344) rats, which are known to differ in their behavioral sensitivities to several drugs of abuse. Morphine 106-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 23153991-7 2013 Immunostaining of c-Fos was very low or absent in the control animals and was consistently up-regulated by morphine, especially in the LC and NTS of the F344 rats and the NAC of the Lewis rats. Morphine 107-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 23306170-0 2013 Soft coral-derived lemnalol alleviates monosodium urate-induced gouty arthritis in rats by inhibiting leukocyte infiltration and iNOS, COX-2 and c-Fos protein expression. lemnalol 19-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-150 23283335-9 2013 Dorsal mPFC light delivery attenuated yohimbine-induced Fos immunoreactivity and disrupted neural activity during in vivo electrophysiological recording in awake rats. Yohimbine 38-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 23251272-6 2013 The positive expression levels of FOS in the MVZ in the SAH and SAH + severed-down vagus nerve (SDV) groups were higher than those in the normal control, sham surgery and SDV groups (P<0.01). N-[(4R)-1-(2-fluorophenyl)-4,5,6,7-tetrahydroindazol-4-yl]-2,1-benzoxazole-3-carboxamide 96-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 23113797-0 2013 Unique gene alterations are induced in FACS-purified Fos-positive neurons activated during cue-induced relapse to heroin seeking. Heroin 114-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 23113797-3 2013 Using Fos as a marker of neural activity, we describe distinct molecular alterations induced in activated versus non-activated neurons during cue-induced heroin seeking after prolonged withdrawal. Heroin 154-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 6-9 23514787-3 2013 Moreover, 60 min after formalin injection, preprotachykinin-A, the SP mRNA, and the immediate early gene cFOS were upregulated in the contralateral striatum. Formaldehyde 23-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-109 23431425-8 2013 Nisoxetine (3 mg/kg) also resulted in greater neural activation, as measured by c-Fos immunohistochemistry, in the arcuate nucleus of the hypothalamus in animals exposed to the high-fat diet. nisoxetine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 26317098-0 2013 The mGlu2/3 Receptor Agonists LY354740 and LY379268 Differentially Regulate Restraint-Stress-Induced Expression of c-Fos in Rat Cerebral Cortex. eglumetad 30-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 23222036-0 2013 Neural substrates of amphetamine-induced behavioral sensitization: unconditioned (zero context) and conditioned (switch versus same context) components in c-fos overexpression. Amphetamine 21-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-160 26317098-0 2013 The mGlu2/3 Receptor Agonists LY354740 and LY379268 Differentially Regulate Restraint-Stress-Induced Expression of c-Fos in Rat Cerebral Cortex. LY 379268 43-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 26317098-3 2013 We compared the efficacy of LY354740 and LY379268 in attenuating restraint-stress-induced expression of the immediate early gene c-Fos in the rat prelimbic (PrL) and infralimbic (IL) cortex. eglumetad 28-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 26317098-3 2013 We compared the efficacy of LY354740 and LY379268 in attenuating restraint-stress-induced expression of the immediate early gene c-Fos in the rat prelimbic (PrL) and infralimbic (IL) cortex. LY 379268 41-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 26317098-7 2013 Because both compounds inhibit serotonin 2A receptor (5-HT2AR)-induced c-Fos expression, we hypothesize that LY354740 and LY379268 have different in vivo properties and that 5-HT2AR activation and restraint stress induce c-Fos through distinct mechanisms. eglumetad 109-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 26317098-7 2013 Because both compounds inhibit serotonin 2A receptor (5-HT2AR)-induced c-Fos expression, we hypothesize that LY354740 and LY379268 have different in vivo properties and that 5-HT2AR activation and restraint stress induce c-Fos through distinct mechanisms. eglumetad 109-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 221-226 26317098-7 2013 Because both compounds inhibit serotonin 2A receptor (5-HT2AR)-induced c-Fos expression, we hypothesize that LY354740 and LY379268 have different in vivo properties and that 5-HT2AR activation and restraint stress induce c-Fos through distinct mechanisms. LY 379268 122-130 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 23560056-8 2013 Additionally, increased transcriptional activity (Fos) was demonstrated in nesfatin neurons during "rebound". nesfatin 75-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 23555638-5 2013 The acute visceral pain behaviors, the colon histological changes and the temporal and spatial changes of NK1R-ir structures and Fos expression in the neurons of the DCN were observed in rats following lower colon instillation with 5% formalin. dcn 166-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-132 23555638-9 2013 Meanwhile, formalin instillation induced a biphasic visceral pain response as well as a strong expression of Fos protein in the nuclei of neurons in the DCN. Formaldehyde 11-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-112 23555638-9 2013 Meanwhile, formalin instillation induced a biphasic visceral pain response as well as a strong expression of Fos protein in the nuclei of neurons in the DCN. dcn 153-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-112 23555638-10 2013 Finally, intrathecal treatment with the NK1R antagonist L732138 attenuated the NK1R internalization, Fos expression and visceral nociceptive responses. 3,5-bis(trifluoromethyl)benzyl N-acetyltryptophan 56-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-104 22999971-9 2012 In both age groups acute morphine activated Fos in the vlPAG, and this effect was attenuated by chronic morphine, specifically in the vlPAG at the level of the laterodorsal tegmental nucleus (LDTg). Morphine 25-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 22917527-5 2012 Enhanced Fos expression in the AcbS was previously found in retrospective timing schedules based on conditional discrimination tasks, but not in a single-operandum immediate timing schedule, the fixed-interval peak procedure. acbs 31-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 22963991-0 2012 A food-associated CS activates c-Fos in VTA DA neurons and elicits conditioned approach. Cesium 18-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 22963991-7 2012 In Experiment 1, eating food (exposure to the US) caused a significantly greater number of VTA DA (TH-labeled) cells to express c-Fos than not eating. Thorium 99-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-133 22963991-8 2012 In Experiment 2, CS (light) presentations caused a significant amount of conditioned approach and a significantly greater number of VTA TH-labeled (DA) cells to express c-Fos. Cesium 17-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-174 22936823-12 2012 Furthermore, treatment decreased c-Fos expression in the dorsal horn, but expressional differences between animals treated with carbamazepine plus pregabalin were not significantly different from those treated with single drug. Carbamazepine 128-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 23039920-4 2012 We found that the ventral bed nucleus of the stria terminalis (vBNST) was a major input to the LH orexin cell field that was significantly Fos-activated during cocaine conditioned place preference (CPP). Cocaine 160-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 22827554-7 2012 SDA completely blunted the induction of c-Fos expression by Ex-4 in the hypothalamic paraventricular nucleus. sda 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 23065200-4 2012 In addition, SP significantly enhanced cerebral Bcl-2, c-Fos and decreased Bax, Caspase-3 proteins positive expression. sp 13-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 23065200-6 2012 Our results confirm that SP can play the protective action against cerebral ischemia reperfusion-induced brain injury by regulating cerebral antioxidant enzymes activities, Bcl-2, Bax, c-Fos and Caspase-3 protein positive expression levels and Bcl-2, Bax, TNF-alpha and Caspase-3 mRNA expression. sp 25-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 185-190 22960130-0 2012 Neuroanatomical substrates of the disruptive effect of olanzapine on rat maternal behavior as revealed by c-Fos immunoreactivity. Olanzapine 55-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 23176293-8 2012 Nox2-dependent ROS generation led to activation and up-regulation of the downstream transcriptional factor AP-1 (i.e. c-Fos and c-Jun), which bound to MMP-9 promoter region, and thereby turned on transcription of MMP-9 gene. Reactive Oxygen Species 15-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 23176293-10 2012 CONCLUSIONS: These results demonstrated that in RBA-1 cells, activation of AP-1 (c-Fos/c-Jun) by the PKC-alpha-mediated Nox2/ROS signals is essential for up-regulation of MMP-9 and cell migration enhanced by BK. Reactive Oxygen Species 125-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 23131151-10 2012 c-Fos activity showed through double labeling, that cell types involved in nicotine action were low threshold (LTS) and fast spiking (FS) inter-neurons, which increased in the DA-depleted striatum. Nicotine 75-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 23131151-10 2012 c-Fos activity showed through double labeling, that cell types involved in nicotine action were low threshold (LTS) and fast spiking (FS) inter-neurons, which increased in the DA-depleted striatum. Dopamine 176-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 22960130-7 2012 Acute olanzapine treatment dose-dependently disrupted various components of maternal behavior (e.g., pup retrieval, pup licking, nest building, crouching) and increased c-Fos immunoreactivity in the medial prefrontal cortex (mPFC), nucleus accumbens shell and core (NAs and NAc), dorsolateral striatum (DLSt), ventral lateral septum (LSv), central amygdala (CeA) and ventral tegmental area (VTA), important brain areas generally implicated in the incentive motivation and reward processing. Olanzapine 6-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-174 23033372-9 2012 WAT injection of capsaicin increased plasma renin, angiotensin II, and norepinephrine levels in OH and caused more c-fos expression in paraventricular nucleus in OH than ON and in ON than obesity-resistant or control rats. Capsaicin 17-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 22749946-13 2012 The ethanol-induced reduction in C-fos gene expression in the prefrontal cortex reveals an abnormal activity of these neurons, which may be relevant in the compulsive consumption of ethanol, the control of reward-related areas and the behavioural phenotype of ethanol addiction. Ethanol 182-189 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 22820553-6 2012 In addition, E-55888 diminished CCI-SN-associated increase in c-Fos immunolabeling in superficial laminae of the lumbar dorsal horn and the locus coeruleus, but increased c-Fos immunolabeling in the nucleus tractus solitarius and the parabrachial area in both control and CCI-SN rats. cci-sn 32-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 22820553-6 2012 In addition, E-55888 diminished CCI-SN-associated increase in c-Fos immunolabeling in superficial laminae of the lumbar dorsal horn and the locus coeruleus, but increased c-Fos immunolabeling in the nucleus tractus solitarius and the parabrachial area in both control and CCI-SN rats. CCI 32-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 22867799-5 2012 A large number of c-Fos-positive neurons were observed in rat injected with a mixture of formalin and vehicle, but not in rat treated with a mixture of formalin and HYP-1. Formaldehyde 89-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 22975136-3 2012 In inflammation group, formalin injection produced a bilateral increase in c-Fos at vestibular nucleus with ipsilesional side higher activity. Formaldehyde 23-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 22932782-5 2012 c-Fos was used to measure neuronal activation in the nucleus of the solitary tract and area postrema in response to intragastric infusions of water, sucrose, or Intralipid. Water 142-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 22932782-5 2012 c-Fos was used to measure neuronal activation in the nucleus of the solitary tract and area postrema in response to intragastric infusions of water, sucrose, or Intralipid. Sucrose 149-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 22960202-4 2012 In the control and saline/formalin injected rats, scattered mRFP1 fluorescence in the spinal cord and the PVN was observed at 0 min, though there was little Fos-LI in the same region. Sodium Chloride 19-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-160 22532480-2 2012 To investigate this potential requirement, antisense Fos oligodeoxynucleotide (ODN) was infused locally into PRH cortex to interfere with Fos production. Oligodeoxyribonucleotides 79-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 22960202-4 2012 In the control and saline/formalin injected rats, scattered mRFP1 fluorescence in the spinal cord and the PVN was observed at 0 min, though there was little Fos-LI in the same region. Formaldehyde 26-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-160 22798527-10 2012 RESULTS: Propofol anesthesia resulted in an inhibition of orexinergic neuron activity as demonstrated by the reduced numbers of c-Fos-immunoreactive orexinergic neurons. Propofol 9-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-133 23073793-9 2012 After DATS treatment, the levels of c-fos and c-jun and MDA in the hepatic tissues were significantly lower in group D than in group C (P<0.05). diallyl trisulfide 6-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 23127505-2 2012 For this purpose combination of Fos (a product of the immediate early gene) labeling with nickel intensified diaminobenzidine (DAB-Ni) and two neuropeptides labeled with Alexa488 and Alexa555 fluorescent dyes on cryo-processed 35-40 microm thick free-floating brain sections was selected. Nickel 90-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 23127505-6 2012 Fos was revealed by avidin-biotin-peroxidase (ABC) complex and visualized by diaminobenzidine chromogen containing nickel chloride salt. 4,4'-Dihydrazino-biphenyl 77-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 23127505-6 2012 Fos was revealed by avidin-biotin-peroxidase (ABC) complex and visualized by diaminobenzidine chromogen containing nickel chloride salt. nickel chloride salt 115-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 23127505-13 2012 CONCLUSIONS: The present study clearly demonstrate that the combination of Fos labeling with DAB-Ni and neuropeptides labeled with Alexa488 and Alexa555 on cryo-processed 35-40 microm thick free-floating brain sections is an excellent approach providing further advantages for quick and reproducible triple immuno-staining enabling to compare the activity of at least two phenotypes of neurons on the same section. dab-ni 93-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 22771386-8 2012 An increase in c-Fos-positive labeled cells was found in the caffeine-treated group at P5-P6 within the same areas described above, with the most clear-cut increase seen in the arcuate nucleus. Caffeine 61-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 22609825-2 2012 After 1h home cage deprivation we observed an increase in the number of c-Fos (neuronal activity marker) immunoreactive cells in several brain regions of the olfactory and stress-related areas in normal neonates at 11 days. Hydrogen 6-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 22906232-12 2012 Dapoxetine acute oral administration (300 mg/kg) to rapid ejaculator rats resulted in (i) diminution of ejaculatory performance (lengthened EL and decreased EF); and (ii) modulation of Fos level of expression in hypothalamic and thalamic nuclei of the brain ejaculatory circuit. dapoxetine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 185-188 22643326-4 2012 The results show that 1 and 10mg/kg SB 243213 enhanced equally c-Fos expression in the subthalamic nucleus (STN) and dose-dependently in the striatum and nucleus accumbens core (NAcc). Antimony 36-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 22993446-0 2012 Fos activation of selective afferents to ventral tegmental area during cue-induced reinstatement of cocaine seeking in rats. Cocaine 100-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 22749946-10 2012 Chronic self-administration of ethanol reduced the expression of the C-fos gene 4- to 12-fold and increased expression of the COX-2 (up to 4-fold) and Homer1a genes in the rat prefrontal cortex. Ethanol 31-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 22749946-13 2012 The ethanol-induced reduction in C-fos gene expression in the prefrontal cortex reveals an abnormal activity of these neurons, which may be relevant in the compulsive consumption of ethanol, the control of reward-related areas and the behavioural phenotype of ethanol addiction. Ethanol 4-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 22749946-13 2012 The ethanol-induced reduction in C-fos gene expression in the prefrontal cortex reveals an abnormal activity of these neurons, which may be relevant in the compulsive consumption of ethanol, the control of reward-related areas and the behavioural phenotype of ethanol addiction. Ethanol 182-189 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 22721675-6 2012 Among 11 brain sites examined, extinction training increased c-Fos expression in basolateral amygdala and prelimbic prefrontal cortex of cocaine-cue extinguished rats relative to both control conditions. Cocaine 137-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 22721675-7 2012 In dorsal subiculum and infralimbic prefrontal cortex, extinction training increased c-Fos expression in both cocaine-cue and saline-cue extinguished rats relative to the no-extinction control condition. Cocaine 110-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 22721675-7 2012 In dorsal subiculum and infralimbic prefrontal cortex, extinction training increased c-Fos expression in both cocaine-cue and saline-cue extinguished rats relative to the no-extinction control condition. Sodium Chloride 126-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 22732530-8 2012 CS increased the percentage of TH/FG+ double-labeled neurons expressing c-Fos in the A1 and LC. Cesium 0-2 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 22732530-9 2012 CS also increased the percentage of TH+ neurons expressing c-Fos within the A1 and A2, independent of their projections to the PVN. Cesium 0-2 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 22288611-9 2012 Rosuvastatin reduced hypertrophy significantly via AT(1) Receptor-PKCbeta2/alpha-ERK-c-fos pathway; protected myocardium against apoptosis via Akt-FOXO1, Bcl-2 family and survivin pathways and consequently suppressed the caspase-3 activity. Rosuvastatin Calcium 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 23047036-5 2012 In ROS17/2.8 cells, FGF2 and FSK each increased the gene expression of c-FOS (7.2-fold and 10.7-fold, respectively). Colforsin 29-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 22580374-7 2012 In addition, treatment of SHR with the selective D1 antagonist SCH-23390 significantly reversed both behavioral hyperactivity and elevated Fos expression in the AcC and cerebral cortex. SCH 23390 63-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 22795918-8 2012 Olcegepant (0.6mg/kg, intravenously) significantly reduced the number of c-Fos immunolabeled cells in spinal nucleus of the trigeminal nerve and upregulation of ATF3 transcript (a marker of neuron injury) but not that of interleukin-6 in trigeminal ganglion of CCI-ION rats. olcegepant 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 22915104-5 2012 Cue-induced heroin seeking increased from 1 to 14 d and was accompanied by increased Fos expression in ~12% of OFC neurons. Heroin 12-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 22643326-5 2012 SB 206553 (1-10mg/kg), at 10mg/kg only, enhanced c-Fos expression in STN, striatum (except the dorsomedial part), NAcc, entopeduncular nucleus, substantia nigra pars reticulata (SNr) and compacta (SNc) and ventral tegmental area. SB 206553 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 22643326-6 2012 S32006 induced a similar increase in c-Fos expression in the medial parts of the striatum and NAcc at doses of 1-10mg/kg while it dose-dependently enhanced c-Fos expression in medial parts of the STN and SNr. S32006 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 22643326-6 2012 S32006 induced a similar increase in c-Fos expression in the medial parts of the striatum and NAcc at doses of 1-10mg/kg while it dose-dependently enhanced c-Fos expression in medial parts of the STN and SNr. S32006 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 25624799-2 2012 We found that c-Fos expression in the rat spinal dorsal horn peaked at 7 hours following the 3.00 mug/kg dexmedetomidine injection, while the levels of c-Fos expression following 0.75 and 1.50 mug/kg dexmedetomidine were similar to those in the spinal dorsal horn of normal rats. Dexmedetomidine 105-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 25624799-6 2012 However, dexmedetomidine appears to have neurotoxic effects in the spinal cord because it increased c-Fos expression in the spinal dorsal horn within 7 hours following administration. Dexmedetomidine 9-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 22561731-0 2012 Cholinergic denervation attenuates phencyclidine-induced c-fos responses in rat cortical neurons. Phencyclidine 35-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 22476004-8 2012 Neuroanatomically, both acute and repeated treatment of HAL significantly increased PCP-induced c-Fos expression in the nucleus accumbens shell (NAs) and the ventral tegmental area (VTA), but reduced it in the central amygdaloid nucleus (CeA). Haloperidol 56-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 22476004-8 2012 Neuroanatomically, both acute and repeated treatment of HAL significantly increased PCP-induced c-Fos expression in the nucleus accumbens shell (NAs) and the ventral tegmental area (VTA), but reduced it in the central amygdaloid nucleus (CeA). Phencyclidine 84-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 22476004-9 2012 Acute and repeated CLZ treatment significantly increased PCP-induced c-Fos expression in the ventral part of lateral septal nucleus (LSv) and VTA, but reduced it in the medial prefrontal cortex (mPFC). Clozapine 19-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 22476004-9 2012 Acute and repeated CLZ treatment significantly increased PCP-induced c-Fos expression in the ventral part of lateral septal nucleus (LSv) and VTA, but reduced it in the medial prefrontal cortex (mPFC). Phencyclidine 57-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 22364199-9 2012 Mifepristone antagonized the TH phosphorylation at Ser31 and the expression of c-Fos expression induced by morphine withdrawal. Mifepristone 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 22364199-9 2012 Mifepristone antagonized the TH phosphorylation at Ser31 and the expression of c-Fos expression induced by morphine withdrawal. Morphine 107-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 22613772-6 2012 We show increased H4Ac at the c-Fos promoter at 1 h post-ECS. h4ac 18-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 22580215-7 2012 The intracellular calcium mobilization and tyrosine phosphorylation were early events, which in turn elicited reactive oxygen species production, gene activation of Cdc42 and c-Fos, and ultimately led to beta-hexosaminidase release. Calcium 18-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 175-180 22613730-0 2012 Cocaine-induced c-Fos expression in rats selectively bred for high or low saccharin intake and in rats selected for high or low impulsivity. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 22613730-7 2012 The low reward-seeking phenotypes showed higher cocaine-induced c-Fos expression in several of these regions. Cocaine 48-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 22613730-8 2012 Low saccharin preferring rats showed higher cocaine-induced c-Fos expression in the nucleus accumbens shell, and low impulsive rats showed higher cocaine-induced c-Fos expression in the orbitofrontal cortex and cingulate gyrus 1 area. Cocaine 44-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 22613730-8 2012 Low saccharin preferring rats showed higher cocaine-induced c-Fos expression in the nucleus accumbens shell, and low impulsive rats showed higher cocaine-induced c-Fos expression in the orbitofrontal cortex and cingulate gyrus 1 area. Cocaine 146-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 22613730-9 2012 In addition, both low impulsive and low saccharin rats had higher cocaine-induced c-Fos in the dorsal medial and dorsal lateral caudate putamen. Cocaine 66-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 22534624-2 2012 This study examined Fos protein expression in response to cocaine cues or to novel cues as a control for activation produced by test novelty. Cocaine 58-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-23 22372932-12 2012 administration of N-PCT increased plasma adrenocorticotrophic hormone and corticosterone concentrations and induced the expression of Fos protein, a marker of neuronal activity, in parvocellular CRF neurones. n-pct 18-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-137 22138163-10 2012 Rimonabant pretreatment potentiated LPS-induced hypophagia, body weight loss and Fos-CRF and Fos-TH expressing neurons. Rimonabant 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-84 22453546-3 2012 OBJECTIVE: This study was conducted to investigate the effects of acupuncture on footshock-induced reinstatement of cocaine-seeking and the expression of c-Fos and the transcription factor cAMP response element-binding protein (CREB) in the NAc, used as markers of neuronal activation in conditions of stress-induced reinstatement to cocaine. Cocaine 334-341 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 22723688-6 2012 Yohimbine- and pellet-priming-induced reinstatement was associated with Fos and GFP induction in mPFC; both reinstatement and neuronal activation were minimally affected by ovarian hormones in both c-fos-GFP and wild-type rats. Yohimbine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 22510382-4 2012 To determine the mechanisms of action of U0126 and PD169316, we studies their effect on some intracellular signaling pathways such as Ca(+)/cAMP-response element binding protein (CREB), c-fos, and transcription factors that regulate mitochondrial biogenesis. 2-(4-nitrophenyl)-4-(4-fluorophenyl)-5-(4-pyridinyl)-1H-imidazole 51-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 186-191 22516009-5 2012 In contrast, Fos-IR in brainstem catecholamine neurons decreased after the administration of GLP-2. Catecholamines 33-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 22884642-6 2012 In addition, significantly more sucrose-induced c-Fos positive cells were found in the CeA, but much less in the external lateral subnucleus of the PBN of dietary obese rats. Sucrose 32-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 22898063-7 2012 Electroacupuncture stimulation induced c-Fos expression in the neurons of the dorsal motor nucleus of the vagus nerve (DMV), which was identified by retrograde tracing. (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 119-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 22183481-4 2012 CIH increased CB TNF-alpha and IL-1beta and c-fos-positive neurons in the NTS, enhanced carotid chemosensory and ventilatory hypoxic responses, and produced hypertension. cih 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 22183481-5 2012 Ibuprofen prevented CB cytokine overexpression and CIH-induced increases in c-fos-positive neurons in the NTS, the enhanced hypoxic ventilatory responses and hypertension, but failed to impede the CB chemosensory potentiation. Ibuprofen 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 22183481-5 2012 Ibuprofen prevented CB cytokine overexpression and CIH-induced increases in c-fos-positive neurons in the NTS, the enhanced hypoxic ventilatory responses and hypertension, but failed to impede the CB chemosensory potentiation. cih 51-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 22580215-7 2012 The intracellular calcium mobilization and tyrosine phosphorylation were early events, which in turn elicited reactive oxygen species production, gene activation of Cdc42 and c-Fos, and ultimately led to beta-hexosaminidase release. Tyrosine 43-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 175-180 21948083-6 2012 Furthermore, upregulation of PKCalpha and p-JNK and downregulation of p-ERK and FOS protein levels were observed in the hippocampus of offspring at P0, P7, and P14 from rats exposed to sevoflurane at gestation, but not pregestation. Sevoflurane 185-196 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 21948083-7 2012 In summary, our data suggest that sevoflurane induces developmental neurotoxicity in rats and this may be attributed to the upregulation of PKCalpha and p-JNK and downregulation of p-ERK and FOS protein in the hippocampus. Sevoflurane 34-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 191-194 22290688-4 2012 The neuropeptide Y release level and the number of c-Fos-like immunoreactive neurons in the baclofen group were significantly attenuated, whereas those in the phaclofen group had increased compared to the saline group. Baclofen 92-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 22290688-8 2012 Overall, the results suggest that baclofen treatment block neuropeptide Y release, subsequently lessening c-Fos expression in cuneate neurons and consequently attenuating neuropathic signal transmission to the thalamus. Baclofen 34-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 22554903-5 2012 c-Fos in the PVN and SO nuclei was found to be significantly increased in trained rats 1h post-exercise compared with control and 24h post-exercise groups. Hydrogen 87-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 22634839-7 2012 In vivo results indicated that cantharidin caused rat hemorrhagic cystitis with hematuria via c-Fos and COX-2 overexpression. Cantharidin 31-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 22634839-10 2012 In conclusion, cantharidin induces rat cystitis through c-Fos and COX-2 over-expression and S. baicalensis can prevent the resulting hematuria because of its anti-inflammatory effects. Cantharidin 15-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 22403798-9 2012 Fos expression after 10% O(2) was similar whether arterial pressure was allowed to decrease (-13 +- 1 mmHg) or was held constant. o(2) 25-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 22167272-4 2012 The results showed that in rats subjected to acute restraint stress, rose essential oil inhalation significantly inhibited the increase in plasma CORT and reduced the increases in the number of c-Fos-positive cells in PVN. rose essential oil 69-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 194-199 22306243-7 2012 Sensory CGRP depletion by capsaicin exacerbated hypoxia-induced pulmonary hypertension in rats, as shown by an increase in right ventricle systolic pressure, mean pulmonary artery pressure and vascular hypertrophy, accompanied with decreased p27 expression and increased expression of phosphorylated ERK1/2, c-fos and c-myc. Capsaicin 26-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 308-313 22405689-2 2012 To gain more understanding of the neural basis of this phenomenon, the current study examined whether a novel taste (0.5% saccharin) supports a different pattern of c-Fos expression than the same taste when it is familiar. Saccharin 122-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 22241629-7 2012 A significant dose-dependent induction in reactive oxygen species (ROS) and early response markers (c-Fos, c-Jun, and GAP-43) were observed in cells exposed to 4-HNE (10, 25, and 50 microM) for 1h. 4-hydroxy-2-nonenal 160-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 22241629-7 2012 A significant dose-dependent induction in reactive oxygen species (ROS) and early response markers (c-Fos, c-Jun, and GAP-43) were observed in cells exposed to 4-HNE (10, 25, and 50 microM) for 1h. Hydrogen 194-196 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 22543127-5 2012 Sucrose solution intake induced significantly more c-Fos and MOR/Fos double-labeled neurons in the CeA than distilled water intake. Sucrose 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 22543127-5 2012 Sucrose solution intake induced significantly more c-Fos and MOR/Fos double-labeled neurons in the CeA than distilled water intake. Sucrose 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 22038537-0 2012 MDMA induces Per1, Per2 and c-fos gene expression in rat suprachiasmatic nuclei. N-Methyl-3,4-methylenedioxyamphetamine 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 22104138-9 2012 Systemic SR142948A administration prevented c-fos mRNA induction in mesolimbic terminal fields (prefrontal cortex, lateral septum, NAcc, ventral subiculum) induced by all three psychostimulants implicating the VTA as the site for NT modulation of stimulant-induced PPI disruption. SR 142948A 9-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 22266344-0 2012 Regional c-Fos and FosB/DeltaFosB expression associated with chronic methamphetamine self-administration and methamphetamine-seeking behavior in rats. Methamphetamine 69-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 22266344-0 2012 Regional c-Fos and FosB/DeltaFosB expression associated with chronic methamphetamine self-administration and methamphetamine-seeking behavior in rats. Methamphetamine 109-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 22306243-11 2012 These results suggest that CGRP inhibits hypoxia-induced proliferation of PASMCs via ERK1/2/p27/c-fos/c-myc pathway. pasmcs 74-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 22236039-7 2012 RESULTS: As expected, icv alcohol stimulated ACTH secretion from the pituitary and Fos expression in the paraventricular nucleus of the hypothalamus (PVN). icv alcohol 22-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 22236039-10 2012 Propranolol also blunted icv alcohol-induced PVN Fos expression. Propranolol 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 22236039-10 2012 Propranolol also blunted icv alcohol-induced PVN Fos expression. Alcohols 29-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 22401907-4 2012 We demonstrate that GLP-1-immunoreactive fibers and fiber terminals topographically overlap with activated Fos-positive neurons in the DMHv in refed rats. bis(N,N-dimethylhydroxamido)hydroxooxovanadate 135-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 22401907-5 2012 Using immunocytochemistry and in situ hybridization histochemistry, we demonstrated GLP-1 receptors in Fos-expressing neurons of the DMH. Dimenhydrinate 133-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-106 22860427-8 2012 RESULTS: Intrathecal administration of nNOS inhibitor 7-Ni and iNOS inhibitors AG decreased the scores of morphine withdrawal, attenuated morphine withdrawal-induced allodynia and also inhibited the increase of Fos protein expression in the spinal cord of morphine withdrawal rats. 7-nitroindazole 54-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 211-214 22414858-0 2012 The alpha4beta2 nicotine acetylcholine receptor agonist ispronicline induces c-Fos expression in selective regions of the rat forebrain. ispronicline 56-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 22414858-6 2012 The restricted distribution of c-Fos to these areas, all of which are directly or indirectly involved in acute stress regulation after a single dose of ispronicline, supports earlier studies that the alpha4beta2 receptors are strongly involved in nicotine-dependent activation of the hypothalamo-pituitary adrenocortical axis. ispronicline 152-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 22414858-6 2012 The restricted distribution of c-Fos to these areas, all of which are directly or indirectly involved in acute stress regulation after a single dose of ispronicline, supports earlier studies that the alpha4beta2 receptors are strongly involved in nicotine-dependent activation of the hypothalamo-pituitary adrenocortical axis. Nicotine 247-255 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 22377455-1 2012 To explore the response to nociceptive stimuli in spinal cord of immature rat and observe the electrical stimulation of the sciatic nerve on synaptic plasticity of the spinal dorsal horn and spinal c-fos expression in rats of different ages, MK-801 was added to the spinal cord of rats, and the resulting changes in field potential as well as c-fos expression were recorded. Dizocilpine Maleate 242-248 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 198-203 22081168-8 2012 In contrast, formalin-induced Fos does not co-localize with TIP39. Formaldehyde 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 22120186-3 2012 Therefore, this study investigated the expression of Fos protein, a marker of neuronal activity, in the brain of rats submitted to FST and treated with the preferential nNOS inhibitor, 7-nitroindazole (7-NI), or with classical antidepressant drugs (Venlafaxine and Fluoxetine). 7-nitroindazole 185-200 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 22120186-3 2012 Therefore, this study investigated the expression of Fos protein, a marker of neuronal activity, in the brain of rats submitted to FST and treated with the preferential nNOS inhibitor, 7-nitroindazole (7-NI), or with classical antidepressant drugs (Venlafaxine and Fluoxetine). 7-nitroindazole 202-206 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 22120186-3 2012 Therefore, this study investigated the expression of Fos protein, a marker of neuronal activity, in the brain of rats submitted to FST and treated with the preferential nNOS inhibitor, 7-nitroindazole (7-NI), or with classical antidepressant drugs (Venlafaxine and Fluoxetine). Venlafaxine Hydrochloride 249-260 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 22120186-3 2012 Therefore, this study investigated the expression of Fos protein, a marker of neuronal activity, in the brain of rats submitted to FST and treated with the preferential nNOS inhibitor, 7-nitroindazole (7-NI), or with classical antidepressant drugs (Venlafaxine and Fluoxetine). Fluoxetine 265-275 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 22340139-7 2012 In immunoreactive oxytocin neurones in the SON and PVN, finasteride increased IL-1beta-induced Fos expression in pregnant rats. Finasteride 56-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 22205547-8 2012 Protracted withdrawal from self-administration enhanced the survival of SGZ BrdU cells, and methamphetamine seeking during protracted withdrawal enhanced Fos expression in the dentate gyrus and medial prefrontal cortex in LgA rats to a greater extent than in ShA and I-ShA rats. Methamphetamine 92-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-157 22431094-0 2012 7-Nitroindazole potentiates c-fos expression induced by muscle tendon vibration in the spinal cord. 7-nitroindazole 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 22431094-1 2012 INTRODUCTION: Expression of c-fos initiated by muscle proprioceptive signaling was studied in rats after inhibition of neuronal nitric oxide synthase (nNOS) with administration of 7-nitroindazole (7-NI). 7-nitroindazole 180-195 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 22431094-1 2012 INTRODUCTION: Expression of c-fos initiated by muscle proprioceptive signaling was studied in rats after inhibition of neuronal nitric oxide synthase (nNOS) with administration of 7-nitroindazole (7-NI). 7-nitroindazole 197-201 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 22431094-2 2012 METHODS: Fos-immunoreactive (Fos-ir) neurons were visualized immunohistochemically in the lumbar cord after vibration of the Achilles tendon and/or 7-NI systemic injections. 7-nitroindazole 148-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 22431094-2 2012 METHODS: Fos-immunoreactive (Fos-ir) neurons were visualized immunohistochemically in the lumbar cord after vibration of the Achilles tendon and/or 7-NI systemic injections. 7-nitroindazole 148-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 22431094-3 2012 RESULTS: The total number of Fos-ir interneurons and motoneurons (per slice) was significantly greater in the 7-NI-pretreated and tendon-vibrated (7-NI + Tv) group than in the isolated tendon vibration group (Tv group). 7-nitroindazole 110-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 22431094-5 2012 CONCLUSIONS: Suppression of NO release after introduction of 7-NI was associated with potentiation of Fos immunoreactivity induced by muscle proprioceptive signaling within distinctive regions of the spinal cord. 7-nitroindazole 61-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 22764597-0 2012 [Effects of electroacupuncture intervention at different time in A day on expression of tyrosine hydroxylase and C-fos in nucleus accumbens in ketamine addiction rats]. Ketamine 143-151 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 22764597-9 2012 CONCLUSION: EA of "Zusanli" (ST 36) and "Sanyinjiao" (SP 6) at 11:00 and 17:00 of the daytime can down-regulate ketamine addiction induced increase of expression of TH and c-fos in the NAc in ketamine addiction rats, which may contribute to its effect in relieving ketamine addiction symptoms in clinic. Ketamine 112-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-177 22764597-9 2012 CONCLUSION: EA of "Zusanli" (ST 36) and "Sanyinjiao" (SP 6) at 11:00 and 17:00 of the daytime can down-regulate ketamine addiction induced increase of expression of TH and c-fos in the NAc in ketamine addiction rats, which may contribute to its effect in relieving ketamine addiction symptoms in clinic. Ketamine 192-200 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-177 22764597-9 2012 CONCLUSION: EA of "Zusanli" (ST 36) and "Sanyinjiao" (SP 6) at 11:00 and 17:00 of the daytime can down-regulate ketamine addiction induced increase of expression of TH and c-fos in the NAc in ketamine addiction rats, which may contribute to its effect in relieving ketamine addiction symptoms in clinic. Ketamine 192-200 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-177 22266344-6 2012 The results showed that acute METH produced a characteristic signature of Fos expression with elevations in striatal, cortical, and extended amygdala regions. Methamphetamine 30-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 22266344-7 2012 Importantly, rats with a 3-week history of METH self-administration displayed similar regional Fos expression to rats receiving METH for the first time. Methamphetamine 43-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 22266344-8 2012 Rats seeking, but not receiving, METH on the test day had augmented Fos in the lateral hypothalamus, septum, and vertical limb of the diagonal band of Broca, suggesting a primary role for these regions in METH-seeking behavior. Methamphetamine 33-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 22266344-12 2012 Overall, these results show persistent upregulated regional brain Fos and FosB/DeltaFosB expression with chronic METH self-administration and indicate a role for the lateral hypothalamus and lateral septum in METH-seeking behavior. Methamphetamine 113-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 22285885-0 2012 Neonatal alcohol exposure and the hippocampus in developing male rats: effects on behaviorally induced CA1 c-Fos expression, CA1 pyramidal cell number, and contextual fear conditioning. Alcohols 9-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 22285885-4 2012 We then examined the relationship between CPFE impairment, hippocampal cell loss, and c-Fos expression in rats exposed to alcohol over PD 4-9 (Experiment 2). Alcohols 122-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 22285885-11 2012 In Experiment 1, c-Fos positive (c-Fos+) cells in the dentate gyrus (DG) were consistently high among rats preexposed to Context A (Pre), Context B (No Pre), or sacrificed directly from their home cage (Home) and did not differ across CPFE phases. cpfe 235-239 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 22285885-16 2012 In addition, lower levels of c-Fos+ cells in alcohol-exposed rats following preexposure may be related to general reductions in the number of CA1 pyramidal cells in these rats. Alcohols 45-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 22457508-3 2012 We found that neurons in rostral LS (LSr) that project to LH are Fos-activated in proportion to cocaine CPP, and that inhibition of LSr neurons with local baclofen and muscimol microinjection (0.3/0.03 nmol) blocks expression of Fos in LH orexin cells and cocaine preference. Muscimol 168-176 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 21995495-0 2012 Mephedrone (4-methylmethcathinone, "meow"): acute behavioural effects and distribution of Fos expression in adolescent rats. mephedrone 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 22305141-2 2012 Expression of c-Fos protein and pERK is mediated by the excitatory neurotransmitter, glutamate. Glutamic Acid 85-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 22305141-9 2012 Microinjection of NMDA or AMPA into the lateral ventricle increased the expression of c-Fos protein in the bilateral MVN of conscious intact labyrinthine rats. N-Methylaspartate 18-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 21995495-0 2012 Mephedrone (4-methylmethcathinone, "meow"): acute behavioural effects and distribution of Fos expression in adolescent rats. mephedrone 12-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 21995495-9 2012 Patterns of Fos expression with mephedrone resembled a combination of those observed with methamphetamine and MDMA, with particularly strong Fos expression in the cortex, dorsal and ventral striatum, ventral tegmental area (typical of both MDMA and methamphetamine) and supraoptic nucleus (typical of MDMA). mephedrone 32-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-15 21995495-9 2012 Patterns of Fos expression with mephedrone resembled a combination of those observed with methamphetamine and MDMA, with particularly strong Fos expression in the cortex, dorsal and ventral striatum, ventral tegmental area (typical of both MDMA and methamphetamine) and supraoptic nucleus (typical of MDMA). N-Methyl-3,4-methylenedioxyamphetamine 110-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-15 21967278-8 2012 Co-administration of caffeine with MDMA increased p-CREB, p-DARPP-32 and c-fos expression when compared with either treatment alone. Caffeine 21-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 21967278-8 2012 Co-administration of caffeine with MDMA increased p-CREB, p-DARPP-32 and c-fos expression when compared with either treatment alone. N-Methyl-3,4-methylenedioxyamphetamine 35-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 22048468-0 2012 Conditioned response evoked by nicotine conditioned stimulus preferentially induces c-Fos expression in medial regions of caudate-putamen. Nicotine 31-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 22155611-3 2012 A Fos immunohistochemistry study recently demonstrated that peripheral OXY administration reduced METH-induced Fos expression within the nucleus accumbens (NAc) core and subthalamic nucleus (STh) in rats. Methamphetamine 98-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 2-5 22155611-3 2012 A Fos immunohistochemistry study recently demonstrated that peripheral OXY administration reduced METH-induced Fos expression within the nucleus accumbens (NAc) core and subthalamic nucleus (STh) in rats. Methamphetamine 98-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 22290557-9 2012 In the TCC, the increase in formalin-induced Fos immunoreactivity was significantly attenuated in the memantine group (p < 0.01). Formaldehyde 28-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 22290557-9 2012 In the TCC, the increase in formalin-induced Fos immunoreactivity was significantly attenuated in the memantine group (p < 0.01). Memantine 102-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 22500249-13 2012 OAB-induced c-Fos and NOS expressions were suppressed by tamsulosin treatment. Tamsulosin 57-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 21110986-6 2012 To explore the neural substrates mediating the observed behavioral effects, we examined the influence of TTA-A2 on amphetamine-induced c-fos expression as well as basal and stimulant-evoked dopamine and glutamate release in the nucleus accumbens. Amphetamine 115-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 21110986-7 2012 TTA-A2 decreased amphetamine-induced c-fos expression as well as MK-801-induced, but not basal, glutamate levels in the nucleus accumbens. Amphetamine 17-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 22048468-3 2012 The purpose of this experiment was to use neurohistochemical analysis of rapidly developing c-Fos protein to elucidate neurobiological loci involved in the processing of nicotine as an interoceptive conditioned stimulus (CS). Nicotine 170-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 22048468-8 2012 In concordance with previous reports, nicotine induced c-Fos expression in the majority of areas tested; however, learning-dependent expression was specific to dorsomedial and ventromedial regions of caudate-putamen (dmCPu, vmCPu). Nicotine 38-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 22048468-9 2012 Only rats in the nicotine-CS condition, when challenged with nicotine, had higher c-Fos expression in the dmCPu and vmCPu. Nicotine 17-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 22048468-9 2012 Only rats in the nicotine-CS condition, when challenged with nicotine, had higher c-Fos expression in the dmCPu and vmCPu. Cesium 26-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 22048468-9 2012 Only rats in the nicotine-CS condition, when challenged with nicotine, had higher c-Fos expression in the dmCPu and vmCPu. Nicotine 61-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 22048468-9 2012 Only rats in the nicotine-CS condition, when challenged with nicotine, had higher c-Fos expression in the dmCPu and vmCPu. dmcpu 106-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 22611921-0 2012 [Effects of polychlorinated biphenyl on the expression of c-fos and c-jun in mesenchymal cells of rats]. Polychlorinated Biphenyls 12-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 21221824-4 2012 We also compared the pattern of Fos expression induced after central injection of a control solution or 8-OHDPAT. 8-Hydroxy-2-(di-n-propylamino)tetralin 104-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 21221824-10 2012 This restricted pattern of Fos expression likely identified the neural substrate responsible for the enhancement of UAM respiratory activity observed after 8-OHDPAT injection. 8-Hydroxy-2-(di-n-propylamino)tetralin 156-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 22133520-7 2012 With regard to brain Fos immunoreactivity, we observed that Furosemide-sodium depletion in the PM-Na group induced a higher number of activated cells in the subfornical organ, ventral subdivision of the paraventricular nucleus and vasopressinergic neurons of the supraoptic nucleus than in the PM-Ctrol animals. Furosemide 60-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 22133520-7 2012 With regard to brain Fos immunoreactivity, we observed that Furosemide-sodium depletion in the PM-Na group induced a higher number of activated cells in the subfornical organ, ventral subdivision of the paraventricular nucleus and vasopressinergic neurons of the supraoptic nucleus than in the PM-Ctrol animals. Sodium 71-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 22178676-8 2012 However, numbers of c-Fos-LI neurons in the glycine and strychnine groups were both significantly less than that in the saline group. Glycine 44-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 22178676-8 2012 However, numbers of c-Fos-LI neurons in the glycine and strychnine groups were both significantly less than that in the saline group. Strychnine 56-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 22138197-2 2012 cytidine-5"-diphosphate (CDP)-choline administration on the activation of oxytocin and vasopressin neurons in the supraoptic (SON) and paraventricular nuclei (PVN), using the immunohistochemical identification of c-Fos expression as a marker of neuronal activation and to correlate this with the plasma hormone levels. Cytidine Diphosphate 0-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-218 22138197-2 2012 cytidine-5"-diphosphate (CDP)-choline administration on the activation of oxytocin and vasopressin neurons in the supraoptic (SON) and paraventricular nuclei (PVN), using the immunohistochemical identification of c-Fos expression as a marker of neuronal activation and to correlate this with the plasma hormone levels. Choline 30-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-218 22138197-8 2012 Dual immunohistochemistry for c-Fos and oxytocin or vasopressin revealed that CDP-choline activates these neurons in a dose-dependent manner. Cytidine Diphosphate Choline 78-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 22138197-9 2012 Light microscopic analyses showed that, about 41%, 75% or 87% of the oxytocin neurons and about 18%, 46% or 82% of the vasopressin neurons in SON express c-Fos, thus activated, by the dosages of 0.5, 1.0 or 2.0 g/kg CDP-choline, respectively. Cytidine Diphosphate Choline 216-227 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 22138197-10 2012 Increases in c-Fos expression were about 29%, 62% or 81% for the oxytocin neurons and about 38%, 70% or 78% for the vasopressin neurons in PVN with the dosages of 0.5, 1.0 or 2.0 g/kg CDP-choline, respectively. Cytidine Diphosphate Choline 184-195 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 22138197-12 2012 In the PVN most of the magnocellular neurons were activated while less number of parvocellular neurons expressed c-Fos in response to CDP-choline challenge. Cytidine Diphosphate Choline 134-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 22138197-13 2012 In correlation with c-Fos data, CDP-choline increased plasma oxytocin and vasopressin levels both dose- and time-dependently. Cytidine Diphosphate Choline 32-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 22081704-6 2012 Intracerebroventricular pretreatment (5 days earlier) with pooled small interfering RNAs targeting p44/42 MAPK reduced total and p-p44/42 MAPK, aldosterone-induced c-Fos expression in the PVN, and the aldosterone-induced increases in RSNA, HR, and MBP. Aldosterone 144-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-169 22611921-1 2012 OBJECTIVE: To study the effects of polychlorinated biphenyl (PCB126) on the expression of c-fos and c-jun in mesenchymal cells (MSCs) of rats. Polychlorinated Biphenyls 35-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 22611921-1 2012 OBJECTIVE: To study the effects of polychlorinated biphenyl (PCB126) on the expression of c-fos and c-jun in mesenchymal cells (MSCs) of rats. 3,4,5,3',4'-pentachlorobiphenyl 61-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 22611921-6 2012 The positive rates of c-fos expression in the low-and high-dose groups were 54.6% and 51.3% at 12 h, and 83.2% and 73.0% at 24 h. The expression of c-fos mRNA detected by RT-PCR was upregulated at 30 min and 1h in PCB126 groups. Hydrogen 208-210 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 22611921-6 2012 The positive rates of c-fos expression in the low-and high-dose groups were 54.6% and 51.3% at 12 h, and 83.2% and 73.0% at 24 h. The expression of c-fos mRNA detected by RT-PCR was upregulated at 30 min and 1h in PCB126 groups. Hydrogen 208-210 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 22611921-6 2012 The positive rates of c-fos expression in the low-and high-dose groups were 54.6% and 51.3% at 12 h, and 83.2% and 73.0% at 24 h. The expression of c-fos mRNA detected by RT-PCR was upregulated at 30 min and 1h in PCB126 groups. 3,4,5,3',4'-pentachlorobiphenyl 214-220 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 22611921-6 2012 The positive rates of c-fos expression in the low-and high-dose groups were 54.6% and 51.3% at 12 h, and 83.2% and 73.0% at 24 h. The expression of c-fos mRNA detected by RT-PCR was upregulated at 30 min and 1h in PCB126 groups. 3,4,5,3',4'-pentachlorobiphenyl 214-220 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 22611921-8 2012 The expression of c-fos and c-jun protein was upregulated in the low-and high-dose groups at 12 h and 24 h. CONCLUSION: PCB126 could promote the proliferation of MSCs and upregulate the expression of cancer associated gene c-fos and c-jun. 3,4,5,3',4'-pentachlorobiphenyl 120-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 22611921-8 2012 The expression of c-fos and c-jun protein was upregulated in the low-and high-dose groups at 12 h and 24 h. CONCLUSION: PCB126 could promote the proliferation of MSCs and upregulate the expression of cancer associated gene c-fos and c-jun. 3,4,5,3',4'-pentachlorobiphenyl 120-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 223-228 22611921-9 2012 The effect of PCB126 on the function of MSCs might be associated with the abnormal expression of c-fos and c-jun gene. 3,4,5,3',4'-pentachlorobiphenyl 14-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 21929659-0 2012 Chronic morphine increases Fos-positive neurons after concurrent cornea and tail stimulation. Morphine 8-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 21929659-1 2012 OBJECTIVE: The aim of the present study was to examine the effect of chronic morphine exposure on diffuse noxious inhibitory controls in a large population of neurons throughout the medullary dorsal horn, as assessed using immunocytochemistry for c-Fos protein. Morphine 77-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 249-252 21929659-4 2012 METHODS: In urethane anesthetized rats, Fos-positive neurons were quantified in chronic morphine and vehicle-treated animals following 52 C noxious thermal stimulation of the cornea with and without the application of a spatially remote noxious stimulus (placement of the tail in 55 C water). Morphine 88-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 21929659-5 2012 RESULTS: When compared to chronic morphine-treated animals that did not receive the spatially remote noxious stimulus, chronic morphine-treated animals given corneal stimulation along with the spatially remote noxious stimulus demonstrated a 163% increase (P < .05) in the number of Fos-positive neurons in the superficial laminae of the medullary dorsal horn and a 682% increase (P < .01) in deep laminae that was restricted to the side ipsilateral to the applied stimulus. Morphine 127-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 286-289 22063717-0 2012 Brain pattern of histone H3 phosphorylation after acute amphetamine administration: its relationship to brain c-fos induction is strongly dependent on the particular brain area. Amphetamine 56-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 21948527-6 2012 Antisense vectors inhibited formalin-evoked c-Fos expression in the spinal cord, indicating decreased nociceptive activity of spinal neurons. Formaldehyde 28-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 22063717-3 2012 Thus, in the present work we studied in adult male rats the effects of a high dose of amphetamine on brain pattern of histone H3 phosphorylation in serine 10 (pH3S(10)) and c-fos expression. Amphetamine 86-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 173-178 22048464-0 2012 Modulation of fronto-cortical activity by modafinil: a functional imaging and fos study in the rat. Modafinil 42-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 22063717-6 2012 Amphetamine increased c-fos expression in medial prefrontal cortex (mPFC), dorsal striatum, nucleus accumbens (Acb), major Island of Calleja (ICjM), central amygdala (CeA), bed nucleus of stria terminalis lateral dorsal (BSTld) and paraventricular nucleus of the hypothalamus (PVN). Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 22455809-5 2012 RESULTS: The relative quantity of c-fos protein and apoptotic index in CIH groups were significantly higher than that of the control group on the 2nd, 4th, 6th and 8th weeks (F were 44.52, 57.56, 24.20 and 13.18, P < 0.05), and these were higher obviously in 5% CIH group than that in 10% CIH group (P < 0.05). cih 71-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 22198527-9 2012 The expression of c-fos in lumbosacral spinal cord dorsal horn (SCDH) was increased significantly by administration of 17beta-estradiol but not estriol, and not by estriol replacement in OVX rats. Estradiol 119-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 22198527-9 2012 The expression of c-fos in lumbosacral spinal cord dorsal horn (SCDH) was increased significantly by administration of 17beta-estradiol but not estriol, and not by estriol replacement in OVX rats. Estriol 144-151 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 22198527-9 2012 The expression of c-fos in lumbosacral spinal cord dorsal horn (SCDH) was increased significantly by administration of 17beta-estradiol but not estriol, and not by estriol replacement in OVX rats. Estriol 164-171 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 22177665-4 2012 Following EIS at 50Hz, a high number of c-Fos positive nuclei were observed showing only marginal tonotopic order in ipsilateral AVCN, in DCN bilaterally, and in contralateral CIC. dcn 138-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 22177665-6 2012 By contrast, the population of neurons expressing c-Fos in DCN and CIC revealed a transient maximum at 73 min. dcn 59-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 22455809-5 2012 RESULTS: The relative quantity of c-fos protein and apoptotic index in CIH groups were significantly higher than that of the control group on the 2nd, 4th, 6th and 8th weeks (F were 44.52, 57.56, 24.20 and 13.18, P < 0.05), and these were higher obviously in 5% CIH group than that in 10% CIH group (P < 0.05). cih 265-268 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 22455809-5 2012 RESULTS: The relative quantity of c-fos protein and apoptotic index in CIH groups were significantly higher than that of the control group on the 2nd, 4th, 6th and 8th weeks (F were 44.52, 57.56, 24.20 and 13.18, P < 0.05), and these were higher obviously in 5% CIH group than that in 10% CIH group (P < 0.05). cih 265-268 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 22455809-6 2012 The expression of c-fos protein and apoptotic index in two CIH groups was different depending upon the different degree and duration of chronic intermittent hypoxia. cih 59-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 22455809-9 2012 The correlation between the relative quantity of c-fos protein and apoptotic index in two CIH groups was positive (r were 0.816 and 0.701, P < 0.01). cih 90-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 22260364-7 2012 GnRH and Fos co-labeling, a marker of GnRH activation, following ovariectomy and hormone priming was reduced by approximately half at all doses; the magnitude of which was not as large as with EB or what we have previously observed with the ERalpha agonist PPT. 4,4',4''-(4-propyl-((1)H)-pyrazole-1,3,5-triyl) tris-phenol 257-260 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 22031786-7 2012 Intravenous GLP-1 induced comparable increases of hindbrain c-Fos immunoreactivity in intact, capsaicin-treated, and vagotomized rats. Capsaicin 94-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 21889521-0 2012 Area postrema lesions attenuate LiCl-induced c-Fos expression correlated with conditioned taste aversion learning. Lithium Chloride 32-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 21889521-2 2012 Systemic administration of LiCl induces c-Fos immunoreactivity in several brain regions, including the AP, nucleus of the solitary tract (NTS), lateral parabrachial nucleus (latPBN), supraoptic nucleus (SON), paraventricular nucleus (PVN), and central nucleus of the amygdala (CeA). Lithium Chloride 27-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 21889521-4 2012 Significant c-Fos induction after LiCl was observed in the CeA and SON of AP-lesioned rats, demonstrating activation independent of an intact AP. Lithium Chloride 34-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 21889521-5 2012 LiCl-induced c-Fos was significantly attenuated in the NTS, latPBN, PVN and CeA of AP-lesioned rats, suggesting that these regions are dependent on AP activation. Lithium Chloride 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 21889521-7 2012 Because c-Fos induction in several regions was correlated with magnitude of CTA acquisition, these regions are implicated in the central mediation of lithium effects during CTA learning. Lithium 150-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-13 22403566-9 2012 Immunofluorescence localization of c-Fos and glutamate revealed that approximately one third of the c-Fos-labeled vestibular neurons showed intense glutamate-like immunofluorescence, far in excess of the stain reflecting the metabolic pool of cytoplasmic glutamate. Glutamic Acid 45-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 22403566-9 2012 Immunofluorescence localization of c-Fos and glutamate revealed that approximately one third of the c-Fos-labeled vestibular neurons showed intense glutamate-like immunofluorescence, far in excess of the stain reflecting the metabolic pool of cytoplasmic glutamate. Glutamic Acid 148-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 22403566-9 2012 Immunofluorescence localization of c-Fos and glutamate revealed that approximately one third of the c-Fos-labeled vestibular neurons showed intense glutamate-like immunofluorescence, far in excess of the stain reflecting the metabolic pool of cytoplasmic glutamate. Glutamic Acid 148-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 22094385-0 2012 Dizocilpine and cycloheximide prevent inhibition of c-Fos gene expression by delta sleep-inducing peptide in the paraventricular nucleus of the hypothalamus in rats with different resistance to emotional stress. Dizocilpine Maleate 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 22094385-0 2012 Dizocilpine and cycloheximide prevent inhibition of c-Fos gene expression by delta sleep-inducing peptide in the paraventricular nucleus of the hypothalamus in rats with different resistance to emotional stress. Cycloheximide 16-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 22094385-1 2012 The effects of the non-competitive NMDA-receptor blocker MK-801 (dizocilpine) and the protein synthesis inhibitor cycloheximide on the delta sleep-inducing peptide (DSIP) inhibition of c-Fos immediate early gene expression were studied in the parvocellular subdivision of the hypothalamic paraventricular nucleus (pPVN) of male Wistar rats with either high or low resistance to emotional stress, predicted from differences in their open-field behaviour. Dizocilpine Maleate 65-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 185-190 22094385-1 2012 The effects of the non-competitive NMDA-receptor blocker MK-801 (dizocilpine) and the protein synthesis inhibitor cycloheximide on the delta sleep-inducing peptide (DSIP) inhibition of c-Fos immediate early gene expression were studied in the parvocellular subdivision of the hypothalamic paraventricular nucleus (pPVN) of male Wistar rats with either high or low resistance to emotional stress, predicted from differences in their open-field behaviour. Cycloheximide 114-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 185-190 23093008-3 2012 Since the mPFC is considered to exert a top-down regulatory control of subcortical brain regions, we examined the effects of local infusion of the NMDAR antagonist, MK801, into the mPFC on the expression of c-Fos protein (widely used marker of neuronal activation) in several subcortical structures. Dizocilpine Maleate 165-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 207-212 22729110-5 2012 Moreover, the increase in c-fos mRNA expression on the formalin-injected side was less pronounced in rats administered landiolol than in saline administered controls. Formaldehyde 55-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 22729110-5 2012 Moreover, the increase in c-fos mRNA expression on the formalin-injected side was less pronounced in rats administered landiolol than in saline administered controls. landiolol 119-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 23093008-5 2012 Bilateral administration of MK801 to the mPFC produced changes in the behavior (increased stereotypy and decreased sleep-like behavior) and complex changes in c-Fos protein expression with significant increases observed in the nucleus accumbens (core and shell), amygdala (basolateral and central nuclei), the CA1 field of the hippocampus, and mediodorsal and paraventricular thalamic nuclei, as compared to the saline group. Dizocilpine Maleate 28-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-164 22729110-5 2012 Moreover, the increase in c-fos mRNA expression on the formalin-injected side was less pronounced in rats administered landiolol than in saline administered controls. Sodium Chloride 137-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 22759963-6 2012 Another transcription factor involved in phosphate-dependent FN synthesis is the AP1 family member c-Fos. Phosphates 41-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 22061687-1 2012 This study investigated the antinociceptive effect of intrathecal escin and examined its effect on the formalin-induced activation of c-Fos and phosphorylated p38 mitogen-activated protein kinase (p-p38 MAPK) in the rat spinal cord. Escin 66-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-139 22061687-1 2012 This study investigated the antinociceptive effect of intrathecal escin and examined its effect on the formalin-induced activation of c-Fos and phosphorylated p38 mitogen-activated protein kinase (p-p38 MAPK) in the rat spinal cord. Formaldehyde 103-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-139 22061687-3 2012 The expression of c-Fos and p-p38 MAPK in the dorsal horn of the spinal cord was detected in the control and escin (40mug) groups using immunohistochemical techniques. Escin 109-114 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 22061687-5 2012 In addition, immunohistochemical experiments showed that the expression of c-Fos and p-p38 MAPK in the spinal cord dorsal horn increased after an injection of formalin into the paw. Formaldehyde 159-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 22061687-6 2012 Interestingly, the 40mug dose of intrathecal escin, which was the larger of the two doses that blocked formalin-induced hyperalgaesia, attenuated the formalin-induced increases in c-Fos and p-p38 MAPK in the dorsal horn of the spinal cord. Escin 45-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 180-185 22061687-6 2012 Interestingly, the 40mug dose of intrathecal escin, which was the larger of the two doses that blocked formalin-induced hyperalgaesia, attenuated the formalin-induced increases in c-Fos and p-p38 MAPK in the dorsal horn of the spinal cord. Formaldehyde 150-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 180-185 22061687-7 2012 The decrease in pain-related behaviours and c-Fos expression indicated that escin produced antinociceptive effects in the rat formalin test. Escin 76-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 22127815-9 2012 Moreover, the capsaicin-induced increase of spinal Fos-immunoreactive cells was significantly diminished by pretreatment, but not posttreatment with Nav1.8 blockers. Capsaicin 14-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 22759963-7 2012 In summary we show that even mildly elevated serum phosphate levels can induce synthesis of the interstitial matrix protein fibronectin through activation of ERK1/2 and AKT signaling pathways in kidney fibroblasts and that the synthesis of fibronectin is mediated by a transcriptional complex consisting of NFATc1, osterix and c-Fos. Phosphates 51-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 327-332 22329817-7 2012 Notably, the sensitivity of oxytocinergic neurons to nicotine was found to be different in the PVN and SON, because only about 55% of the SON oxytocinergic neurons co-stored Fos even after the highest dose of nicotine. Nicotine 53-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 174-177 22087024-8 2012 This mechanism was supported by glucocorticoid injection immediately before a psychological stress (30 min, white noise); methylprednisolone caused dose-dependent attenuation of stress-induced corticosterone release and expression of the activity marker c-fos mRNA in the paraventricular nucleus but did not block the pituitary response to CRH. Methylprednisolone 122-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 254-259 22550540-5 2012 Treatment of EA at BL60 significantly inhibited flinching behavior and c-fos expression induced by formalin injection into the paw, compared to a control group. Formaldehyde 99-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 21892689-5 2012 Finally, 4 weeks after MNT, pre-stimulation administration of L-NAME (N (omega)-Nitro-L: -arginine methyl ester) or 7-NI (7-nitroindazole) suppressed the amount of NPY release from the stimulated terminals and thus attenuated c-Fos expression in the CN. NG-Nitroarginine Methyl Ester 70-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 226-231 22675357-4 2012 A significant decrease in the number of Fos-like immunoreactive (LI) cells was observed in the CCI + lidocaine AC IOP group compared with that in the CCI group. CCI 95-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 22675357-4 2012 A significant decrease in the number of Fos-like immunoreactive (LI) cells was observed in the CCI + lidocaine AC IOP group compared with that in the CCI group. Lidocaine 101-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 22675357-4 2012 A significant decrease in the number of Fos-like immunoreactive (LI) cells was observed in the CCI + lidocaine AC IOP group compared with that in the CCI group. CCI 150-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 21892689-0 2012 Nitric oxide implicates c-Fos expression in the cuneate nucleus following electrical stimulation of the transected median nerve. Nitric Oxide 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 21892689-5 2012 Finally, 4 weeks after MNT, pre-stimulation administration of L-NAME (N (omega)-Nitro-L: -arginine methyl ester) or 7-NI (7-nitroindazole) suppressed the amount of NPY release from the stimulated terminals and thus attenuated c-Fos expression in the CN. NG-Nitroarginine Methyl Ester 62-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 226-231 22107704-10 2012 C-Fos/GABA double-fluorescence clearly confirmed a SCS-induced activation of GABA-immunoreactive cells in responders immediately after SCS. gamma-Aminobutyric Acid 77-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 23090268-0 2012 Comparison of tibolone and 17beta-estradiol administration on the expression of zonula occludens-1, occludin, glial fibrillary acidic protein and c-fos levels in the brain cortex and hippocampus of female rats. Estradiol 27-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-151 23090268-1 2012 OBJECTIVES: To compare the effect exerted by oral tibolone or intramuscular 17beta-estradiol administration on the expression of ZO-1, occludin, GFAP and c-fos levels in the brain cortex and hippocampus of ovariectomized rats. tibolone 50-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 23090268-1 2012 OBJECTIVES: To compare the effect exerted by oral tibolone or intramuscular 17beta-estradiol administration on the expression of ZO-1, occludin, GFAP and c-fos levels in the brain cortex and hippocampus of ovariectomized rats. Estradiol 76-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 22074881-0 2012 Distribution of c-Fos immunoreactivity in the rat brain following abuse-like toluene vapor inhalation. Toluene 77-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 22074881-3 2012 Thus in the present study we systematically mapped in over 140 brain structures the distribution of c-Fos immunoreactivity (c-Fos IR), a proxy for neural activation, following exposure to an abuse-like concentration (~5000 ppm) of toluene vapor for 0, 5, 10 or 30 min. Toluene 231-238 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 22074881-3 2012 Thus in the present study we systematically mapped in over 140 brain structures the distribution of c-Fos immunoreactivity (c-Fos IR), a proxy for neural activation, following exposure to an abuse-like concentration (~5000 ppm) of toluene vapor for 0, 5, 10 or 30 min. Toluene 231-238 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 22074881-4 2012 Quantitative analyses revealed increases in c-Fos IR in about one-third of the brain structures examined, with most of these structures significantly activated only after prolonged toluene exposure. Toluene 181-188 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 22074881-9 2012 The pattern of c-Fos IR evoked by inhalation of toluene vapor appears distinct from other psychoactive substances, consistent with the unique and complex behavioral outcomes associated with acute toluene inhalation. Toluene 48-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 22074881-9 2012 The pattern of c-Fos IR evoked by inhalation of toluene vapor appears distinct from other psychoactive substances, consistent with the unique and complex behavioral outcomes associated with acute toluene inhalation. Toluene 196-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 22162685-5 2012 Lidocaine pretreatment also reduced the number of injury-induced NPY-like immunoreactive (NPY-LI) fibers and c-Fos-LI neurons within the CN in a dose-dependent manner. Lidocaine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 23071721-7 2012 In addition, CP-154,526 reduced the number of TH containing neurons expressing c-Fos in the VTA after naloxone-precipitated morphine withdrawal. cp-154 13-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 23071721-7 2012 In addition, CP-154,526 reduced the number of TH containing neurons expressing c-Fos in the VTA after naloxone-precipitated morphine withdrawal. Naloxone 102-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 23071721-7 2012 In addition, CP-154,526 reduced the number of TH containing neurons expressing c-Fos in the VTA after naloxone-precipitated morphine withdrawal. Morphine 124-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 22916276-8 2012 In the medial prefrontal cortex, we evidenced a negative correlation between Fos expression in its dorsal part and open arm-induced freezing in NaCl-treated rats but not in cocaine withdrawn rats. Sodium Chloride 144-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-80 22905122-4 2012 RESULTS: Tungstate treatment reduces the expression of gluconeogenic enzymes (PEPCK, G6Pase, and FBPase) and also regulates transcription factors accountable for the control of hepatic metabolism (c-jun, c-fos and PGC1alpha). tungstate 9-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 204-209 22808090-7 2012 Unilateral administration of either baclofen or muscimol (220 pmol) induced similar patterns of c-fos immunoreactivity in several hypothalamic sites but differed in its induction in the central nucleus of the amygdala. Baclofen 36-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 22808090-7 2012 Unilateral administration of either baclofen or muscimol (220 pmol) induced similar patterns of c-fos immunoreactivity in several hypothalamic sites but differed in its induction in the central nucleus of the amygdala. Muscimol 48-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 22590628-7 2012 Importantly, SB334867 attenuated the somatic symptoms of withdrawal, and reduced morphine withdrawal-induced c-Fos expression in the nucleus accumbens (NAc) shell, bed nucleus of stria terminalis, central amygdala and hypothalamic paraventricular nucleus, but did not modify the HPA axis activity. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 22590628-7 2012 Importantly, SB334867 attenuated the somatic symptoms of withdrawal, and reduced morphine withdrawal-induced c-Fos expression in the nucleus accumbens (NAc) shell, bed nucleus of stria terminalis, central amygdala and hypothalamic paraventricular nucleus, but did not modify the HPA axis activity. Morphine 81-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 21863235-0 2012 Pre-treatment with the mGlu2/3 receptor agonist LY379268 attenuates DOI-induced impulsive responding and regional c-Fos protein expression. LY 379268 48-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 21863235-8 2012 DOI enhanced Fos IR within fronto-cortical and limbic brain structures, and this effect was blocked by LY379268 pre-treatment. LY 379268 103-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 21863235-9 2012 Double immunofluorescence labeling showed a specific co-localization of DOI-elicited Fos IR with GABAergic (GAD(67)-positive) cells lacking the calcium-binding protein parvalbumin while LY379268 increased Fos IR in GABAergic and non-GABAergic cells. LY 379268 186-194 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 21892689-5 2012 Finally, 4 weeks after MNT, pre-stimulation administration of L-NAME (N (omega)-Nitro-L: -arginine methyl ester) or 7-NI (7-nitroindazole) suppressed the amount of NPY release from the stimulated terminals and thus attenuated c-Fos expression in the CN. 7-nitroindazole 116-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 226-231 21892689-5 2012 Finally, 4 weeks after MNT, pre-stimulation administration of L-NAME (N (omega)-Nitro-L: -arginine methyl ester) or 7-NI (7-nitroindazole) suppressed the amount of NPY release from the stimulated terminals and thus attenuated c-Fos expression in the CN. 7-nitroindazole 122-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 226-231 21957165-7 2011 In addition, hindbrain SB334867 induced c-Fos in the nucleus of the solitary tract. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 23-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 22490213-8 2012 Intracisterna magna pretreatment of p38MAPK inhibitor SB203580 resulted in potent attenuation of phase II of pain behavior (P < 0.05), while the expression of c-fos was also inhibited, especially at the point of 120 min (P < 0.01). SB 203580 54-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 162-167 22359483-0 2011 c-Fos Expression in the Nucleus of the Solitary Tract in Response to Salt Stimulation in Rats. Salts 69-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 22359483-7 2011 From this study, salt stimulation with high concentration (1.0 M NaCl) induced significantly higher c-Fos expression in intermediate NST and dorsal-medial and dorsal-middle subregions of the NST compared to distilled water stimulation. Salts 17-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 22359483-7 2011 From this study, salt stimulation with high concentration (1.0 M NaCl) induced significantly higher c-Fos expression in intermediate NST and dorsal-medial and dorsal-middle subregions of the NST compared to distilled water stimulation. Sodium Chloride 65-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 21889971-0 2011 Spinal glycinergic and GABAergic neurons expressing C-fos after capsaicin stimulation are increased in rats with contralateral neuropathic pain. Capsaicin 64-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 21964377-7 2011 ICV CRF also increased Fos-positive non-CRF neurons in the CeA and the BNSTov, which was inhibited by AS-30 in both areas, and inhibited by antalarmin in the BNSTov only. antisauvagine 30 102-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 21889971-2 2011 We have investigated this phenomenon by studying the activation pattern (using C-fos labeling) of spinal glycinergic and GABAergic (Gly/GABA) neurons after capsaicin injection in the ipsilateral hind paw of rats that were preconditioned with an acute or chronic pain stimulus in the contralateral hind paw or rats that were not preconditioned (control). Capsaicin 156-165 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 21889971-5 2011 We found that after capsaicin injection in rats preconditioned with CFA inflammation (4 days), sham-SNI or with SNI neuropathic pain, the numbers (27 +- 3, 21 +- 2, and 21 +- 2, respectively) and percentages (55% +- 4, 43% +- 2, and 42% +- 2, respectively) of C-fos activated neurons that were Gly/GABA increased significantly as compared with control (10 +- 1 and 25% +- 2). Capsaicin 20-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 260-265 21889971-6 2011 The increase in the total number of C-fos activated Gly/GABA neurons was present primarily in the superficial dorsal horn (laminae I and II; control: 9%; CFA 4 days: 56%; SNI 2 weeks: 42%). Glycine 52-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 21889971-6 2011 The increase in the total number of C-fos activated Gly/GABA neurons was present primarily in the superficial dorsal horn (laminae I and II; control: 9%; CFA 4 days: 56%; SNI 2 weeks: 42%). gamma-Aminobutyric Acid 56-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 21889971-7 2011 This increase in C-fos activation of Gly/GABA neurons occurred without significant changes in the total number of C-fos activated neurons, and without any significant changes in the mechanical thresholds in the hind paws after capsaicin injection. Glycine 37-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 21889971-7 2011 This increase in C-fos activation of Gly/GABA neurons occurred without significant changes in the total number of C-fos activated neurons, and without any significant changes in the mechanical thresholds in the hind paws after capsaicin injection. gamma-Aminobutyric Acid 41-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 21889971-8 2011 The results showed that one-sided chronic pain, especially inflammation, significantly increases the C-fos activation pattern of spinal Gly/GABA neurons on the other side of the spinal cord. Glycine 136-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 21889971-8 2011 The results showed that one-sided chronic pain, especially inflammation, significantly increases the C-fos activation pattern of spinal Gly/GABA neurons on the other side of the spinal cord. gamma-Aminobutyric Acid 140-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 21930190-0 2011 Induction of Fos expression in the rat forebrain after intragastric administration of monosodium L-glutamate, glucose and NaCl. Sodium Glutamate 86-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 21958872-4 2011 In addition, only one intrathecal injection of tachykinin NK1 receptor siRNA reduced carrageenan-induced inflammation and thermal hyperalgesia significantly and markedly attenuated the induction of flinching after formalin injection and c-Fos expression in the dorsal horn following formalin injection. Carrageenan 85-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 237-242 21930190-0 2011 Induction of Fos expression in the rat forebrain after intragastric administration of monosodium L-glutamate, glucose and NaCl. Glucose 110-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 21930190-0 2011 Induction of Fos expression in the rat forebrain after intragastric administration of monosodium L-glutamate, glucose and NaCl. Sodium Chloride 122-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 21930190-3 2011 In the present study, we mapped activation sites of the rat forebrain after intragastric load of 60 mM monosodium l-glutamate (MSG) by measurement of Fos protein, a functional marker of neuronal activation. Sodium Glutamate 103-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-153 21930190-6 2011 On the other hand, glucose administration exclusively enhanced Fos induction in the nucleus accumbens. Glucose 19-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 21930190-7 2011 Both MSG and glucose enhanced Fos induction in three brain regions (the habenular nucleus, paraventricular nucleus, and central nucleus of the amygdala). Glucose 13-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 22054645-8 2011 Finally, inhibition of PKCzeta/PKMzeta activity decreased the expression of Fos in response to formalin and CFA in both superficial and deep laminae of the dorsal horn. Formaldehyde 95-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 22114293-7 2011 CS presentation outside the extinction context renewed conditional freezing and was associated with significantly more c-fos expression in BA-projecting neurons in the VH and PL than that induced by CS presentation in the extinction context. Cesium 0-2 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 21821064-5 2011 First exposure to stress in late diestrus evoked a 50% decrease in Fos expression compared to non-stressed controls, particularly in the lateral and dorsolateral sectors of the rostral PAG. pag 185-188 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 21821064-6 2011 In contrast, in experienced rats in late diestrus the pattern of Fos expression increased up to 4-fold, particularly in the ventral half of the caudal PAG but also in the lateral and dorsolateral parts. pag 151-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 22000083-17 2011 The number of c-Fos-labeled cells in the PAG was generally low but there was a reliable increase in c-Fos expression in dorsolateral PAG (dlPAG) following the SR test in the brains of rats that went through the EU-EXT procedure as compared with those that either went through the more-traditional CSO extinction procedure or experienced no extinction at all. dlpag 138-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 22001573-0 2011 Sustained Fos expression is observed in the developing brainstem auditory circuits of kanamycin-treated rats. Kanamycin 86-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-13 22001573-2 2011 As c-fos was expected to be expressed in the SOC by kanamycin-induced cochlear damage, the expression of c-fos protein (Fos) was investigated using immunohistochemistry in kanamycin-treated rat pups. Kanamycin 52-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 5-8 22001573-2 2011 As c-fos was expected to be expressed in the SOC by kanamycin-induced cochlear damage, the expression of c-fos protein (Fos) was investigated using immunohistochemistry in kanamycin-treated rat pups. Kanamycin 172-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 5-8 22001573-2 2011 As c-fos was expected to be expressed in the SOC by kanamycin-induced cochlear damage, the expression of c-fos protein (Fos) was investigated using immunohistochemistry in kanamycin-treated rat pups. Kanamycin 172-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-118 22001573-2 2011 As c-fos was expected to be expressed in the SOC by kanamycin-induced cochlear damage, the expression of c-fos protein (Fos) was investigated using immunohistochemistry in kanamycin-treated rat pups. Kanamycin 172-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-123 22001573-4 2011 In contrast, in kanamycin-treated rats, Fos-IR was consistently observed through P9. Kanamycin 16-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 22001573-5 2011 Because a significant increase in terminal uridine deoxynucleotidyl transferase-mediated 2"-deoxyuridine 5"-triphosphate-biotin nick-end labeling (TUNEL) and glial fibrillary acidic protein (GFAP) IR was not demonstrated in the MNTB and LSO of kanamycin-treated rats, the increased Fos-IR does not appear to indicate an ongoing pathologic process, but may be related to the increased activity caused by the disturbance in excitatory and inhibitory balance between brainstem auditory circuits. Uridine 43-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 282-285 22001573-5 2011 Because a significant increase in terminal uridine deoxynucleotidyl transferase-mediated 2"-deoxyuridine 5"-triphosphate-biotin nick-end labeling (TUNEL) and glial fibrillary acidic protein (GFAP) IR was not demonstrated in the MNTB and LSO of kanamycin-treated rats, the increased Fos-IR does not appear to indicate an ongoing pathologic process, but may be related to the increased activity caused by the disturbance in excitatory and inhibitory balance between brainstem auditory circuits. Biotin 121-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 282-285 21978215-6 2011 Intraperitoneal injection of diazepam, an anxiolytic agent, but not indomethacin, an antipyretic, significantly reduced both the stress-induced hyperthermia and Fos expression in these medullary raphe neuronal populations. Diazepam 29-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-164 22034979-6 2011 We found a strong correlation between the expression of food anticipation measured by running-wheel activity and Fos expression levels in the DMH of ABA-normal rats, whereas no correlation was found in ABA-random rats. 1,2-Dimethylhydrazine 142-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-116 22034979-6 2011 We found a strong correlation between the expression of food anticipation measured by running-wheel activity and Fos expression levels in the DMH of ABA-normal rats, whereas no correlation was found in ABA-random rats. alisol B 23-acetate 149-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-116 21903328-8 2011 The reduction was accompanied by a decrease in formalin-induced expression of spinal c-Fos protein that serves as an index of spinal nociceptive processing. Formaldehyde 47-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 22082421-0 2011 Increase of capsaicin-induced trigeminal Fos-like immunoreactivity by 5-HT(7) receptors. Capsaicin 12-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 22082421-8 2011 RESULTS: Capsaicin but not its vehicle induced Fos-like immunoreactivity within laminae I and II of trigeminal nucleus caudalis. Capsaicin 9-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 22082421-10 2011 Interestingly, capsaicin-induced Fos-like immunoreactivity was abolished by SB-656104 pretreatment thus suggesting involvement of endogenous 5-HT. Capsaicin 15-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 22082421-10 2011 Interestingly, capsaicin-induced Fos-like immunoreactivity was abolished by SB-656104 pretreatment thus suggesting involvement of endogenous 5-HT. SB-656104 76-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 21902723-9 2011 In SHR, the number of c-Fos-positive neurons was significantly lower following the administration of prazosin compared with vehicle. Prazosin 101-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 21875950-10 2011 Oxycodone treatment significantly increased the number of c-fos-positive neurons in the area postrema and NTS compared with water controls. Oxycodone 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 21875950-11 2011 As expected, cisplatin also increased the number of c-fos-positive cells in these regions. Cisplatin 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 21855627-0 2011 c-Fos expression after deep brain stimulation of the pedunculopontine tegmental nucleus in the rat 6-hydroxydopamine Parkinson model. Oxidopamine 99-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 21781979-8 2011 In the supraoptic nucleus (SON) of sham rats, water deprivation significantly increased Fos staining while water intake following dehydration prevented this increase. Water 46-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-91 21689994-1 2011 Pilocarpine (cholinergic muscarinic agonist) injected peripherally may act centrally to produce pressor responses; in the present study, using c-fos immunoreactive expression, we investigated the forebrain and brainstem areas activated by pressor doses of intravenous (i.v.) Pilocarpine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-148 21781979-9 2011 Water deprivation significantly increased Fos staining in the SON of SLNX rats. Water 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 21781979-10 2011 Following water intake after 46 h water deprivation in SLNX rats, Fos staining in the ipsilateral SON was significantly greater than the contralateral SON and significantly lower than 48 h water deprivation. Water 10-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 21781979-10 2011 Following water intake after 46 h water deprivation in SLNX rats, Fos staining in the ipsilateral SON was significantly greater than the contralateral SON and significantly lower than 48 h water deprivation. Water 34-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 21781979-10 2011 Following water intake after 46 h water deprivation in SLNX rats, Fos staining in the ipsilateral SON was significantly greater than the contralateral SON and significantly lower than 48 h water deprivation. Water 34-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 21781979-11 2011 In the nucleus of the solitary tract (NTS) of sham rats, both water deprivation and water intake produced significant increases in Fos staining bilaterally compared to euhydrated controls. Water 62-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-134 21781979-11 2011 In the nucleus of the solitary tract (NTS) of sham rats, both water deprivation and water intake produced significant increases in Fos staining bilaterally compared to euhydrated controls. Water 84-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-134 21781979-12 2011 In SLNX rats, water deprivation significantly increased Fos in both ipsilateral and contralateral NTS that was not different from sham rats. Water 14-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 21784133-3 2011 Neurons in both regions constitutively express the transcription factor Forkhead protein2 (FoxP2), and become c-Fos activated after prolonged sodium depletion. Sodium 142-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 21781979-13 2011 SLNX significantly decreased Fos staining in the ipsilateral NTS of rats given access to water after dehydration compared to the corresponding sham treated rats. Water 89-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 21781979-15 2011 This suggests that the superior laryngeal nerve contributes to changes in Fos staining in the NTS and SON following water intake in dehydrated rats. Water 116-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 20734160-0 2011 Gabapentin completely attenuated the acute morphine-induced c-Fos expression in the rat nucleus accumbens. Gabapentin 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 21803943-9 2011 ICV misoprostol increased the c-Fos immunoreactivity of PVN neurons, an effect that was substantially reduced by the EP(3) receptor antagonist. icv 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 21803943-9 2011 ICV misoprostol increased the c-Fos immunoreactivity of PVN neurons, an effect that was substantially reduced by the EP(3) receptor antagonist. Misoprostol 4-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 21087552-6 2011 At comparative doses, aripiprazole distinguished itself from the other PAs by causing the least rotation in the hypo-dopaminergic model (indicating the least intrinsic activity) and showed the highest Fos expression in the nucleus accumbens (indicating functional D2 antagonism). Aripiprazole 22-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 201-204 20734160-0 2011 Gabapentin completely attenuated the acute morphine-induced c-Fos expression in the rat nucleus accumbens. Morphine 43-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 20734160-3 2011 Therefore, we examined the combined effects of GBP-morphine on acute morphine-induced c-Fos expression in rat nucleus accumbens. Morphine 69-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 20734160-14 2011 Present results showed that GBP-morphine combination action prevented the acute morphine-induced c-Fos expression in rat nucleus accumbens. Morphine 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 20734160-14 2011 Present results showed that GBP-morphine combination action prevented the acute morphine-induced c-Fos expression in rat nucleus accumbens. Morphine 80-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 21641003-9 2011 The increase in c-fos immunoreactivity occurred in DRN neurons immunopositive for the GABA marker parvalbumin. gamma-Aminobutyric Acid 86-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 21778018-5 2011 Systemic PGB also significantly reduced Fos labelling in lumbosacral spinal cords of rats given noxious repetitive CRD; however, PGB did not alter this measure of neural activity in the brainstem. Pregabalin 9-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 22097274-1 2011 The objective of this study was to investigate the changes of the behaviors, EEG and expression of c-fos, c-jun on induced seizure in rats by injecting penicillin after transmitting epileptic discharge from brain tissue to muscular tissue on skull. Penicillins 152-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 21969878-10 2011 On the other hand, chronic ethanol-binge increased mRNA expression of angiotensinogen and c-fos. Ethanol 27-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 21718761-0 2011 Sucrose modifies c-fos mRNA expression in the brain of rats maintained on feeding schedules. Sucrose 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 21718761-6 2011 The present study investigated how adding palatable sucrose to feeding schedules affects the pattern of brain c-fos mRNA expression during FAA (0-3 h) and 1 h following feeding. Sucrose 52-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 21718761-8 2011 Adding sucrose to scheduled feeding displaced c-fos mRNA expression from the dorsomedial and paraventricular hypothalamic nuclei and posterior lateral hypothalamus (LH) to the prefrontal cortex, lateral septum, nucleus accumbens and anterior LH. Sucrose 7-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 21723961-0 2011 Protective action of endogenously generated H2S on hypoxia-induced respiratory suppression and its relation to antioxidation and down-regulation of c-fos mRNA in medullary slices of neonatal rats. Hydrogen Sulfide 44-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 21723961-2 2011 The protective action of endogenous H(2)S and its relation to antioxidation and down-regulation of c-fos mRNA were investigated in the present study. Hydrogen Sulfide 36-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 21723961-5 2011 The hypoxia-induced up-regulation of c-fos mRNA could be markedly antagonized by S-adenosyl-l-methionine (SAM, activator of CBS), but greatly increased by NH(2)OH. S-Adenosylmethionine 81-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 21723961-5 2011 The hypoxia-induced up-regulation of c-fos mRNA could be markedly antagonized by S-adenosyl-l-methionine (SAM, activator of CBS), but greatly increased by NH(2)OH. S-Adenosylmethionine 106-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 21723961-7 2011 These results indicate that endogenously generated H(2)S was involved in protection of the medullary respiratory centers against hypoxic injury partly via antioxidation and down-regulation of c-fos. Hydrogen Sulfide 51-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 192-197 21964712-0 2011 c-Fos expression in rat brainstem following intake of sucrose or saccharin. Sucrose 54-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 21440571-5 2011 Here we examine a neurochemical mechanism underlying this effect and report that GSN stimulated food intake is highly correlated with the induction of the neuronal activation marker cFOS within two nuclei with a demonstrated role in central glucose sensing and appetite, the arcuate nucleus of the hypothalamus (ARC) and lateral hypothalamic area (LHA). Glucosamine 81-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 182-186 21440571-5 2011 Here we examine a neurochemical mechanism underlying this effect and report that GSN stimulated food intake is highly correlated with the induction of the neuronal activation marker cFOS within two nuclei with a demonstrated role in central glucose sensing and appetite, the arcuate nucleus of the hypothalamus (ARC) and lateral hypothalamic area (LHA). Glucose 241-248 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 182-186 21440571-6 2011 Furthermore, GSN stimulated cFOS within the ARC was observed in orexigenic neurons expressing the endogenous melanocortin receptor antagonist agouti-related peptide (AgRP) and neuropeptide Y (NPY), but not those expressing the anorectic endogenous melanocortin receptor agonist alpha-melanocyte stimulating hormone (alpha-MSH). Glucosamine 13-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-32 21440571-7 2011 In the LHA, GSN stimulated cFOS was found within arousal and feeding associated orexin/hypocretin (ORX), but not orexigenic melanin-concentrating hormone (MCH) expressing neurons. Glucosamine 12-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-31 21726175-6 2011 KV treatment also demonstrates significant restoration in ATZ-induced alterations in the expression of apoptosis markers viz., p53, Bax, Bcl2, caspase-3, caspase-9, cyclooxygenase-2 (COX-2), c-Jun and c-fos. kolaviron 0-2 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 201-206 21726175-6 2011 KV treatment also demonstrates significant restoration in ATZ-induced alterations in the expression of apoptosis markers viz., p53, Bax, Bcl2, caspase-3, caspase-9, cyclooxygenase-2 (COX-2), c-Jun and c-fos. atz 58-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 201-206 21792724-6 2011 The results indicated that: (1) significantly more terminal deoxynucleotidyl transferase dUTP nick end labeling-positive cells were observed at 1, 3, and 7 d following injury and (2) the expression of both c-Jun and c-Fos was induced 10 min after incision and had markedly higher levels 2 h post-injury. deoxyuridine triphosphate 89-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 216-221 21689104-7 2011 Moreover, ALCAR enhanced the expression of several memory-associated proteins including c-Fos, synapsin I in rat hippocampus. Acetylcarnitine 10-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 21762292-7 2011 Moreover, increased ethanol consumption was associated with lowered blood corticosterone levels, indicating a blunted HPA signaling, which was reversed by intra-PVN injection of picrotoxin, as indicated by the increased Fos immunostaining-positive cells in the PVN and the increased blood corticosterone levels. Ethanol 20-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 220-223 21762292-7 2011 Moreover, increased ethanol consumption was associated with lowered blood corticosterone levels, indicating a blunted HPA signaling, which was reversed by intra-PVN injection of picrotoxin, as indicated by the increased Fos immunostaining-positive cells in the PVN and the increased blood corticosterone levels. Picrotoxin 178-188 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 220-223 21964712-0 2011 c-Fos expression in rat brainstem following intake of sucrose or saccharin. Saccharin 65-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 21964712-5 2011 These results suggest that the PBN and NST may be involved in sweet taste perception and modulation of sweet tastant intake, but the significantly enhanced intensity of Fos expression induced by sucrose indicates that PBN/NST neuronal activity is driven by the integrated effects of sweet taste sensation and post-ingestive signals. Sucrose 195-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-172 21635907-2 2011 To better characterize this possible mechanism, c-fos immunohistochemistry was first used to determine the effects of systemic administration of the full 5-HT1AR agonist +-8-OH-DPAT on L-Dopa-induced immediate early gene expression within M1 and the prefrontal cortex (PFC) of rats with unilateral medial forebrain bundle (MFB) dopamine (DA) lesions. 8-Hydroxy-2-(di-n-propylamino)tetralin 172-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 21635907-2 2011 To better characterize this possible mechanism, c-fos immunohistochemistry was first used to determine the effects of systemic administration of the full 5-HT1AR agonist +-8-OH-DPAT on L-Dopa-induced immediate early gene expression within M1 and the prefrontal cortex (PFC) of rats with unilateral medial forebrain bundle (MFB) dopamine (DA) lesions. Levodopa 185-191 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 21635907-5 2011 While no treatment effects were seen within the PFC, systemic +-8-OH-DPAT suppressed L-Dopa-induced c-fos within M1. 8-Hydroxy-2-(di-n-propylamino)tetralin 64-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 21635907-5 2011 While no treatment effects were seen within the PFC, systemic +-8-OH-DPAT suppressed L-Dopa-induced c-fos within M1. Levodopa 85-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 21509809-6 2011 RESULTS: Comparison of the saline and BoNT-A treated groups showed a significant decrease in L6 (i.e., 67%, P < 0.001) and S1 (i.e., 47%, P < 0.01) c-fos expression (43%) in BoNT-A treated rats compared to saline controls. Sodium Chloride 27-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 21554894-5 2011 This estrogen-dependent inhibition on induced sodium appetite (approximately 50% reduction) can be correlated with changes in Fos activation observed in the organum vasculosum of the lamina terminalis (OVLT) and DRN, in response to sodium depletion. Sodium 46-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-129 21554894-5 2011 This estrogen-dependent inhibition on induced sodium appetite (approximately 50% reduction) can be correlated with changes in Fos activation observed in the organum vasculosum of the lamina terminalis (OVLT) and DRN, in response to sodium depletion. Sodium 232-238 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-129 21527272-5 2011 COX-2 antagonist (NS-398, 10mg/kg; IP) administration prior to LPS (100mug/kg; IP) prevented anorexia and reduced c-Fos expression the DRN, MnR, nucleus tractus solitarii and several related forebrain areas. N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide 18-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 21340718-11 2011 Enterochromaffin cells and serotonin in colon were related to the elevated c-fos in CNS (P < 0.05). Serotonin 27-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 21860534-7 2011 Valproate+ECS significantly increased the c-Fos protein levels of the prefrontal cortex compared with the ECS group. Valproic Acid 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 21570990-0 2011 Sensitized activation of Fos and brain-derived neurotrophic factor in the medial prefrontal cortex and ventral tegmental area accompanies behavioral sensitization to amphetamine. Amphetamine 166-177 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 21570990-11 2011 Amphetamine challenge increased Fos and BDNF expression in the mPFC regardless of prior drug experience, and further augmented mPFC BDNF expression in sensitized rats. Amphetamine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 21570990-12 2011 Similarly, more Fos-FG and Fos-BDNF double-labeling was observed in the mPFC of sensitized rats compared to drug-naive rats after amphetamine challenge. Amphetamine 130-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 21570990-12 2011 Similarly, more Fos-FG and Fos-BDNF double-labeling was observed in the mPFC of sensitized rats compared to drug-naive rats after amphetamine challenge. Amphetamine 130-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 21570990-13 2011 Repeated amphetamine treatment also increased VTA BDNF, while both acute and repeated amphetamine treatment increased Fos and Fos-BDNF co-labeling, an effect enhanced in sensitized rats. Amphetamine 86-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 21570990-13 2011 Repeated amphetamine treatment also increased VTA BDNF, while both acute and repeated amphetamine treatment increased Fos and Fos-BDNF co-labeling, an effect enhanced in sensitized rats. Amphetamine 86-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-129 21782156-7 2011 Interestingly, at some time-points, combined treatment with morphine and PD168,077 substantially increased c-Fos, FosB/DeltaFosB and P-CREB expression. Morphine 60-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 21782156-7 2011 Interestingly, at some time-points, combined treatment with morphine and PD168,077 substantially increased c-Fos, FosB/DeltaFosB and P-CREB expression. pd168 73-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 21745546-0 2011 MDMA-induced c-Fos expression in oxytocin-containing neurons is blocked by pretreatment with the 5-HT-1A receptor antagonist WAY 100635. N-Methyl-3,4-methylenedioxyamphetamine 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 21745546-9 2011 WAY 100635 significantly reduced MDMA-induced c-Fos expression in a subset of brain regions examined. N-Methyl-3,4-methylenedioxyamphetamine 33-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 21331665-0 2011 Effects of heating coagulation of middle meningeal artery on plasma CGRP level and c-fos expression in migraine rat triggered by nitroglycerin. Nitroglycerin 129-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 21435143-7 2011 RESULTS: Treatment with alpha-LA decreased the 8-OH-dG and Fos expressions to controls" levels, but did not affect CD11b. Thioctic Acid 24-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 21331665-5 2011 We found that NTG led to markedly increase in plasma CGRP level and c-fos expression in trigeminal nucleus caudalis compared with the isotonic saline-treated group (P < 0.05). Nitroglycerin 14-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 21524663-9 2011 Our results reveal that c-fos and glial markers were triggered by the combined stress of non-thermal irradiation and the toxic effect of picrotoxin on cerebral tissues. Picrotoxin 137-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 21365203-2 2011 In rats, the dopamine agonist, apomorphine (APO), reduces PPI and expression of the early gene, c-fos, within the nucleus accumbens (NAC) core. Dopamine 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 21636214-9 2011 Hyperalgesia and c-Fos overexpression were blocked only by prestress treatment with diazepam and post-stress treatment with ketamine, whereas changes in GABA and glutamate release were reversed by prestress treatment with diazepam. Diazepam 84-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 21636214-9 2011 Hyperalgesia and c-Fos overexpression were blocked only by prestress treatment with diazepam and post-stress treatment with ketamine, whereas changes in GABA and glutamate release were reversed by prestress treatment with diazepam. Ketamine 124-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 21365203-2 2011 In rats, the dopamine agonist, apomorphine (APO), reduces PPI and expression of the early gene, c-fos, within the nucleus accumbens (NAC) core. Apomorphine 31-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 21262266-5 2011 None of the drugs reduced HAL-induced Fos expression in the striatum or frontal cortex, and M100,907 actually potentiated HAL-induced Fos expression in the n. accumbens. Haloperidol 122-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-137 21734275-3 2011 Whisker stimulation elicited cerebral blood flow (CBF) increases concomitantly with c-Fos upregulation in pyramidal cells that coexpressed cyclooxygenase-2 (COX-2) and GABA interneurons that coexpressed vasoactive intestinal polypeptide and/or choline acetyltransferase, but not somatostatin or parvalbumin. gamma-Aminobutyric Acid 168-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 21543638-3 2011 Because Fos and FosB are transcription factors involved in activator protein (AP)-1 driven central nervous system neuronal adaptations, this study determined if CIH causes increased Fos or FosB staining in brain regions that regulate SNA and autonomic function. cih 161-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-11 21543638-3 2011 Because Fos and FosB are transcription factors involved in activator protein (AP)-1 driven central nervous system neuronal adaptations, this study determined if CIH causes increased Fos or FosB staining in brain regions that regulate SNA and autonomic function. cih 161-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 21700643-4 2011 RESULTS: We observed tactile allodynia of the face and hindpaws, as well as enhanced Fos expression within the trigeminal nucleus caudalis (TNC) following CSD induced by KCl injection into the cortex, but not by pinprick. Potassium Chloride 170-173 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 21700643-5 2011 Application of KCl onto the dura elicited cutaneous allodynia and increased Fos staining in the TNC but did not elicit CSD events. Potassium Chloride 15-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 21649655-13 2011 In the control group, L-NAME application led to fewer nNOS-immunoreactive cells in the cerebral cortex and TNC, and fewer Fos-immunoreactive cells in the TNC; however, L-NAME was without effect on the CSD pattern. NG-Nitroarginine Methyl Ester 22-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 21433064-8 2011 In addition, fasudil significantly decreased ISO-induced JNK activation, ERK translocation to the nucleus and subsequent c-fos, c-jun expression and upregulated c-FLIP(L) expression. fasudil 13-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-126 21515256-10 2011 Further, the relative amount of Arc mRNA compared to c-Fos mRNA was higher for BDNF, equal for NMDA and lower for AMPA. N-Methylaspartate 95-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 21295078-2 2011 Several lines of evidence have shown that cAMP-response element binding protein (CREB), extracellular signal-regulated kinase (ERK), and c-fos have pivotal role in CPP induced by drugs of abuse, such as morphine, cocaine, nicotine, and alcohol. Morphine 203-211 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 21295078-2 2011 Several lines of evidence have shown that cAMP-response element binding protein (CREB), extracellular signal-regulated kinase (ERK), and c-fos have pivotal role in CPP induced by drugs of abuse, such as morphine, cocaine, nicotine, and alcohol. Cocaine 213-220 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 21295078-2 2011 Several lines of evidence have shown that cAMP-response element binding protein (CREB), extracellular signal-regulated kinase (ERK), and c-fos have pivotal role in CPP induced by drugs of abuse, such as morphine, cocaine, nicotine, and alcohol. Nicotine 222-230 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 21295078-2 2011 Several lines of evidence have shown that cAMP-response element binding protein (CREB), extracellular signal-regulated kinase (ERK), and c-fos have pivotal role in CPP induced by drugs of abuse, such as morphine, cocaine, nicotine, and alcohol. Alcohols 236-243 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 21463663-3 2011 Thirty minutes after APS, induced by a formalin injection into the left hind paw, PVN, BSTov, CeA and npEW all showed a peak in cFos mRNA expression that was followed by a robust increase in cFos protein-immunoreactivity, indicating a rapid increase in (immediate early) gene expression in all four brain nuclei. aps 21-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-132 21463663-3 2011 Thirty minutes after APS, induced by a formalin injection into the left hind paw, PVN, BSTov, CeA and npEW all showed a peak in cFos mRNA expression that was followed by a robust increase in cFos protein-immunoreactivity, indicating a rapid increase in (immediate early) gene expression in all four brain nuclei. aps 21-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 191-195 21463663-3 2011 Thirty minutes after APS, induced by a formalin injection into the left hind paw, PVN, BSTov, CeA and npEW all showed a peak in cFos mRNA expression that was followed by a robust increase in cFos protein-immunoreactivity, indicating a rapid increase in (immediate early) gene expression in all four brain nuclei. Formaldehyde 39-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-132 21463663-3 2011 Thirty minutes after APS, induced by a formalin injection into the left hind paw, PVN, BSTov, CeA and npEW all showed a peak in cFos mRNA expression that was followed by a robust increase in cFos protein-immunoreactivity, indicating a rapid increase in (immediate early) gene expression in all four brain nuclei. Formaldehyde 39-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 191-195 22097732-6 2011 Fos-like positive neurons in dorsal horn of withdrawal group were 380 +/- 71, which were higher than those of U0126 group(287 +/- 54, P < 0.05). U 0126 110-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 21461920-5 2011 We observed that LPS (10 mug/ml) treatment of rat astrocytoma cells, C6, for 24 h significantly increased intracellular Ca(2+) ion and expression of inducible nitric oxide synthase (iNOS), nuclear factor kappa-B (NF-kB), C/EBP homologous protein 10 (CHOP), c-fos, and c-jun proteins. lps 17-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 257-262 21461920-8 2011 Treatment with nimesulide also attenuated LPS-induced Ca(2+) ion, iNOS, NF-kB, and c-fos expressions, but does not significantly influence CHOP, c-jun protein expressions, and mRNA levels of IL-6, IL-1alpha, IL-1beta, and mPGES-1 genes. nimesulide 15-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 21461920-8 2011 Treatment with nimesulide also attenuated LPS-induced Ca(2+) ion, iNOS, NF-kB, and c-fos expressions, but does not significantly influence CHOP, c-jun protein expressions, and mRNA levels of IL-6, IL-1alpha, IL-1beta, and mPGES-1 genes. lps 42-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 21439021-8 2011 The amounts of total and phosphorylated c-Fos increased over time in the presence of ATZ + mercaptosuccinic acid, whereas the amounts of total and phosphorylated c-Jun remained unchanged. Amitrole 85-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 21449042-7 2011 The Cd-exposed group exhibited a degradation of seminiferous tubules and inhibition of a stepwise change in methylation in the coding region of c-fos in testis at day 70 pp. Cadmium 4-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-149 21447066-5 2011 We first observed that alcohol up-regulated c-fos signals in the locus coeruleus, the main noradrenergic brain cell group; and that it activated (nor)adrenergic medullary cells (A1-A2/C1-C3). Alcohols 23-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 19632743-4 2011 Water maze experience significantly increased Fos expression in the suprapyramidal blade and Fos was highest in the septal pole of the dentate gyrus whether the animal learned a platform location, swam in the absence of a platform or remained in their cage. Water 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 19632743-4 2011 Water maze experience significantly increased Fos expression in the suprapyramidal blade and Fos was highest in the septal pole of the dentate gyrus whether the animal learned a platform location, swam in the absence of a platform or remained in their cage. Water 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 21412226-8 2011 However, opposite to our expectations of an inhibitory influence of androgen receptors in the posterior BST, PVN AVP mRNA and stress-induced Fos were augmented in response to DHT and attenuated in response to HF. Dihydrotestosterone 175-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 21334415-6 2011 Moreover, E10x pretreatment attenuated the cocaine-induced increase in Egr1, Egr2, and c-fos expression in the rat frontal cortex, whereas phosphorylation of ERK1/2, one of the representative upstream activators of these genes, increased significantly following cocaine treatment. Cocaine 43-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 21251645-7 2011 Double in situ hybridization experiments revealed that PCP (10 mg/kg intraperitoneal [IP]) markedly increased c-fos expression in glutamatergic neurons of several cortical areas (prefrontal, somatosensory, retrosplenial, entorhinal) and in thalamic nuclei, including MD/CM. Phencyclidine 55-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 110-115 21251645-8 2011 Phencyclidine also increased c-fos expression in the amygdala; yet, it had a small effect in the hippocampus. Phencyclidine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 21251645-10 2011 Clozapine (5 mg/kg IP) had no effect by itself but significantly prevented PCP-induced c-fos expression. Clozapine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 21251645-10 2011 Clozapine (5 mg/kg IP) had no effect by itself but significantly prevented PCP-induced c-fos expression. Phencyclidine 75-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 21459678-0 2011 Modulation of conditioned fear, fear-conditioned analgesia, and brain regional c-Fos expression following administration of muscimol into the rat basolateral amygdala. Muscimol 124-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-84 21459678-2 2011 In this study, we investigated the effects of intra-BLA administration of the GABA(A) receptor agonist muscimol on the expression of conditioned-fear, formalin-evoked nociception, and fear-conditioned analgesia in rats, and the associated alterations in brain regional expression of the immediate early gene product and marker of neuronal activity, c-Fos. Muscimol 103-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 349-354 21459678-5 2011 The suppression of fear behavior by intra-BLA muscimol was associated with increased c-Fos expression in the central nucleus of the amygdala (CeA) and throughout the periaqueductal grey (PAG). Muscimol 46-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 21459678-5 2011 The suppression of fear behavior by intra-BLA muscimol was associated with increased c-Fos expression in the central nucleus of the amygdala (CeA) and throughout the periaqueductal grey (PAG). pag 187-190 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 21459678-6 2011 These intra-BLA muscimol-induced increases in c-Fos expression were abolished in rats receiving intraplantar formalin injection. Muscimol 16-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 21459678-6 2011 These intra-BLA muscimol-induced increases in c-Fos expression were abolished in rats receiving intraplantar formalin injection. Formaldehyde 109-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 21764680-4 2011 RESULTS: Compared with control group, the rats with oral sucrose solution stimulation exhibited significantly increased c-Fos-expressing and double-labeled neurons in the nucleus of the solitary tract (NST), the parabrachial nucleus (PBN) and the amygdala. Sucrose 57-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 21412223-6 2011 Nicotine selectively increased c-fos mRNA expression in the nucleus accumbens shell and basolateral amygdala in adolescent, but not adult animals, and altered serotonin markers in these regions as well as the prefrontal cortex. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 21507338-1 2011 A study was carried out to examine the effects of acute and chronic L-DOPA treatment on the distribution of the immediate-early gene (IEG) proteins (FosB, c-Fos, and Zif268) in forebrain regions in a unilateral 6-hydroxydopamine (6-OHDA) rat model of Parkinson"s disease. Levodopa 68-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-160 21439021-8 2011 The amounts of total and phosphorylated c-Fos increased over time in the presence of ATZ + mercaptosuccinic acid, whereas the amounts of total and phosphorylated c-Jun remained unchanged. 2-thiomalic acid 91-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 21431982-4 2011 Group II: the rats were stimulated as for Group I and c-Fos expression in NTS, DMV and SON was examined. (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 79-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 21418337-9 2011 The difference in c-Fos expression after mating disappeared when the two groups were ovariectomised and received steroid replacement. Steroids 113-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 21431982-7 2011 In Group I, the discharge frequency increased in all the three nuclei, while in Group II, Fos expression in NTS, DMV and SON was, respectively, greater than that of Group III. (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 113-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-93 21326191-6 2011 This effect was not related to differences in the ability to attribute predictive value to a conditioned stimulus (as measured by conditioned approach behavior), but was potentially linked to the development of behavioral supersensitivity to amphetamine and to augmented amphetamine-induced immediate early-gene expression (c-fos and Nur77) in dorsal striatopallidal and striatonigral cells. Amphetamine 242-253 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 324-329 21229349-5 2011 Acute injection of METH increased c-fos, fosB, fra2, junB, Egr1-3, Nr4a1 (Nur77), and Nr4a3 (Nor-1) mRNA levels in the striatum of saline-pretreated rats. Methamphetamine 19-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 21326191-6 2011 This effect was not related to differences in the ability to attribute predictive value to a conditioned stimulus (as measured by conditioned approach behavior), but was potentially linked to the development of behavioral supersensitivity to amphetamine and to augmented amphetamine-induced immediate early-gene expression (c-fos and Nur77) in dorsal striatopallidal and striatonigral cells. Amphetamine 271-282 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 324-329 21559464-5 2011 A single percutaneous IT injection of BoNT-B 0.5 U at 2 or 5 days prior to IPLT formalin reduced NK1-R internalization and C-Fos expression. Formaldehyde 80-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 21376061-9 2011 The number of Fos (+) neurons decreased in acute and chronic L-NAME and decreased in acute L-arginine groups. NG-Nitroarginine Methyl Ester 61-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 21376061-9 2011 The number of Fos (+) neurons decreased in acute and chronic L-NAME and decreased in acute L-arginine groups. Arginine 91-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 21376061-10 2011 Following ENSC, Fos (+) neurons did not change in acute L-NAME but decreased in the chronic L-NAME groups, and decreased in both acute and chronic L-arginine groups. NG-Nitroarginine Methyl Ester 92-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 21376061-10 2011 Following ENSC, Fos (+) neurons did not change in acute L-NAME but decreased in the chronic L-NAME groups, and decreased in both acute and chronic L-arginine groups. Arginine 147-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 21362452-8 2011 The data suggest that vagus nerve stimulation may inhibit heroin- or heroin cue-induced relapse, possibly by regulation of the expression of Fos and CREB in nucleus accumbens. Heroin 58-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 21362452-8 2011 The data suggest that vagus nerve stimulation may inhibit heroin- or heroin cue-induced relapse, possibly by regulation of the expression of Fos and CREB in nucleus accumbens. Heroin 69-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 21486808-5 2011 Moreover, activation of neurons in the CnF induced increased expression of Fos protein in the dorsolateral PAG. pag 107-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 21277949-6 2011 The results show that acute nicotine injection induces Fos expression in 5-HT neurons in a region-specific manner. Nicotine 28-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 21185871-8 2011 Importantly, the increase of corticosterone level after escape or panic-like response was paralleled by an increase of neuronal activation of c-Fos in both the parvocellular and magnocellular paraventricular nucleus of the hypothalamus. Corticosterone 29-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-147 21185871-9 2011 Moreover, the c-Fos data also showed a decrease in the number of positive cells particularly for the ESCIT as well as the BUSP in comparison with the saline stimulated animals. Sodium Chloride 150-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 21080080-0 2011 Effect of ketamine administration on memory consolidation, p-CREB and c-fos expression in the hippocampal slices of minor rats. Ketamine 10-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 23431025-0 2011 Effects of Aripiprazole and Haloperidol on Fos-like Immunoreactivity in the Prefrontal Cortex and Amygdala. Aripiprazole 11-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 23431025-0 2011 Effects of Aripiprazole and Haloperidol on Fos-like Immunoreactivity in the Prefrontal Cortex and Amygdala. Haloperidol 28-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 23431025-2 2011 The aim of this study was to investigate the effects of aripiprazole and haloperidol on c-FOS expression in rat brain. Aripiprazole 56-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 23431025-2 2011 The aim of this study was to investigate the effects of aripiprazole and haloperidol on c-FOS expression in rat brain. Haloperidol 73-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 23431025-9 2011 RESULTS: The administration of aripiprazole at all doses (1, 10 or 30 mg/kg) resulted in greater Fos-like immunoreactivity (FLI) in the investigated brain areas, as compared to the vehicle. Aripiprazole 31-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 23431025-12 2011 In the Ce and BL, a significant increase in Fos-positive neurons was observed only with 0.1 mg/kg of haloperidol. Haloperidol 101-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 20888277-4 2011 First, in order to determine if visceral pain relies on changes in spinal activity, we analyzed Fos expression in the spinal cord of rats treated with butyrate following a challenge with repetitive noxious colorectal distension. Butyrates 151-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-99 20888277-6 2011 In control rats treated with saline, noxious repetitive colorectal distensions evoked Fos expression only in L1-2-6 and S1 spinal segments. Sodium Chloride 29-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-89 21331757-5 2011 In addition, the effect of AEA (administered 30 min before NTG injection) was investigated on NTG-induced Fos expression and evaluated 4 h following NTG injection. aea 27-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 21331757-5 2011 In addition, the effect of AEA (administered 30 min before NTG injection) was investigated on NTG-induced Fos expression and evaluated 4 h following NTG injection. Nitroglycerin 94-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 21331757-5 2011 In addition, the effect of AEA (administered 30 min before NTG injection) was investigated on NTG-induced Fos expression and evaluated 4 h following NTG injection. Nitroglycerin 94-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 21331757-7 2011 Pre-treatment with AEA significantly reduced the NTG-induced neuronal activation in nucleus trigeminalis caudalis, confirming the results obtained in our previous study, and in area postrema, while the same treatment induced an increase of Fos expression in paraventricular and supraoptic nuclei of the hypothalamus, parabrachial nucleus, and periaqueductal grey. aea 19-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 240-243 21331757-7 2011 Pre-treatment with AEA significantly reduced the NTG-induced neuronal activation in nucleus trigeminalis caudalis, confirming the results obtained in our previous study, and in area postrema, while the same treatment induced an increase of Fos expression in paraventricular and supraoptic nuclei of the hypothalamus, parabrachial nucleus, and periaqueductal grey. Nitroglycerin 49-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 240-243 21262543-0 2011 The endocannabinoid arachidonylethanolamide attenuates aspects of lipopolysaccharide-induced changes in energy intake, energy expenditure and hypothalamic Fos expression. Endocannabinoids 4-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-158 21262543-0 2011 The endocannabinoid arachidonylethanolamide attenuates aspects of lipopolysaccharide-induced changes in energy intake, energy expenditure and hypothalamic Fos expression. anandamide 20-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-158 21262543-4 2011 AEA attenuated LPS-induced fever and hypophagia, abolished LPS-induced decreases in Fos expression within the arcuate and ventromedial nucleus of the hypothalamus, while both AEA and LPS independently increased Fos expression within the nucleus accumbens. anandamide 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 21262543-4 2011 AEA attenuated LPS-induced fever and hypophagia, abolished LPS-induced decreases in Fos expression within the arcuate and ventromedial nucleus of the hypothalamus, while both AEA and LPS independently increased Fos expression within the nucleus accumbens. anandamide 175-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 211-214 21080080-9 2011 Therefore, administration of ketamine may temporally affect the ability of memory consolidation rate of minor rats through suppressing the expression of p-CREB and c-fos protein in hippocampus. Ketamine 29-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-169 21199535-6 2011 KEY RESULTS: Baclofen administration was associated with a significant attenuation of the behavioral and EMG responses (at 1 and 3 mg kg(-1)) and expression of Fos in T8 and T9 segments in neonatal iodoacetamide sensitized rats. Baclofen 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-163 21199535-8 2011 Baclofen (3 mg kg(-1)) also significantly reduced the expression of spinal Fos in response to gastric distention in control rats to a lesser extent than sensitized rats. Baclofen 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 21163296-6 2011 Results show that SR141716A activates c-Fos expression in brainstem areas receiving vagal inputs. Rimonabant 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 21125398-10 2011 Furthermore, nicotine increased c-Fos expression in the CeA, but not the medial or basolateral amygdaloid nucleus. Nicotine 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 21125398-11 2011 The increase of c-Fos in the CeA was significantly inhibited by MLA. methyllycaconitine 64-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 21430166-7 2011 Functional analysis by stimulus-evoked c-Fos expression and electrophysiological recording showed that host axons formed active synapses with graft neurons at the injury site with the signal propagating by graft axons to the DCN. dcn 225-228 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 21216264-6 2011 Morphine did not significantly enhance, but suppressed Fos expression in certain brain regions of drug-naive WLP and WHP rats. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 21216264-7 2011 Fos expression revealed considerable differences in the responses of WLP and WHP rats to morphine challenge, particularly after methadone pretreatment. Morphine 89-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 21216264-7 2011 Fos expression revealed considerable differences in the responses of WLP and WHP rats to morphine challenge, particularly after methadone pretreatment. Methadone 128-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 21886557-2 2011 These changes are considered as consequences of cocaine-induced molecular adaptation such as CREB and c-Fos. Cocaine 48-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 21411666-1 2011 Numerous studies with the neural activity marker Fos indicate that cocaine activates only a small proportion of sparsely distributed striatal neurons. Cocaine 67-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 20933023-0 2011 Central injections of noradrenaline induce reinstatement of cocaine seeking and increase c-fos mRNA expression in the extended amygdala. Norepinephrine 22-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 21886557-3 2011 Recently, methanolic extracts from licorice was reported to decrease cocaine-induced dopamine release and c-Fos expression in the nucleus accumbens. methanolic 10-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 21886557-6 2011 In addition, LQ inhibited CREB phosphorylation and c-Fos expression in the striatum and the nucleus accumbens induced by acute cocaine. liquiritigenin 13-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 21886557-6 2011 In addition, LQ inhibited CREB phosphorylation and c-Fos expression in the striatum and the nucleus accumbens induced by acute cocaine. Cocaine 127-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 20817578-7 2011 Furthermore, the MCS increased the Fos-IR in the periaqueductal gray, the anterior cingulate cortex and the central and basolateral amygdaloid nuclei. mcs 17-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 21285311-0 2011 Estradiol alters Fos-immunoreactivity in the hippocampus and dorsal striatum during place and response learning in middle-aged but not young adult female rats. Estradiol 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 21068718-5 2011 Microinjection of recombinant TNFalpha into the vlPAG followed by intraperitoneal naloxone resulted in morphine withdrawal-like behavioral signs, and upregulation of pERK1/2, expression of Fos, and phosphorylation of cAMP response element binding (pCREB) protein. Naloxone 82-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 189-192 20938761-0 2011 Detection of conspecific pheromones elicits fos expression in GABA and calcium-binding cells of the rat vomeronasal system-medial extended amygdala. gamma-Aminobutyric Acid 62-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 20938761-0 2011 Detection of conspecific pheromones elicits fos expression in GABA and calcium-binding cells of the rat vomeronasal system-medial extended amygdala. Calcium 71-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 20938761-4 2011 Likewise, a great colocalization of fos with GABA, calretinin, and calbindin was observed in the vomeronasal system-medial extended amygdala. gamma-Aminobutyric Acid 45-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-39 21168404-6 2011 The transcription factor c-jun is significantly reduced while c-fos becomes up regulated after cAMP elevation. Cyclic AMP 95-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 21160468-4 2011 Bilateral ibotenic acid lesions of the LS increased hypothalamo-pituitary-adrenocortical (HPA) axis responses to forced swim stress indicated by enhanced plasma ACTH and corticosterone responses and higher stress-induced c-Fos-like immunoreactivity in the paraventricular hypothalamic nucleus. Ibotenic Acid 10-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 221-226 21108936-3 2011 In each site, after rats are fed an extremely low-sodium diet for over a week, neurons increase their expression of an activity-induced transcription factor, c-Fos. Sodium 50-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 21146592-7 2011 Dual-labeled immunohistochemistry showed a significant increase in the percentage of CaMKII-positive neurons also immunoreactive for c-Fos in the BLA, CEA and MEA of ferret odor-exposed rats compared to control and butyric acid-exposed groups. cea 151-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 21146592-7 2011 Dual-labeled immunohistochemistry showed a significant increase in the percentage of CaMKII-positive neurons also immunoreactive for c-Fos in the BLA, CEA and MEA of ferret odor-exposed rats compared to control and butyric acid-exposed groups. mea 159-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 21146592-7 2011 Dual-labeled immunohistochemistry showed a significant increase in the percentage of CaMKII-positive neurons also immunoreactive for c-Fos in the BLA, CEA and MEA of ferret odor-exposed rats compared to control and butyric acid-exposed groups. Butyric Acid 215-227 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 20973776-10 2011 Concurrent with preventing reinstatement, SB-334867 decreased Fos in NAc core, PrL and OFC following immediate reinstatement. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 42-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 20973776-11 2011 Following protracted abstinence, SB-334867 treatment decreased reinstatement-induced Fos in the PrL, OFC and piriform cortices. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 33-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 20973776-13 2011 The effects of SB-334867 on both behaviour and Fos expression suggest that the orexin system is implicated in cue-induced reinstatement, although some loci may shift following protracted abstinence. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 15-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 21199267-7 2011 RESULTS: minocycline treatment reversed the mechanical hyperalgesia, increased Fos expression and decreased the KCC2 expression detected in STZ-diabetic rats to control levels. Minocycline 9-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 21107265-0 2011 Effects of intrathecal isoflurane administration on nociception and Fos expression in the rat spinal cord. Isoflurane 23-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 21107265-9 2011 Immunohistochemistry and real-time reverse transcriptase PCR revealed that isoflurane administration inhibited formalin injection-induced c-fos expression in the spinal cord. Isoflurane 75-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 21107265-9 2011 Immunohistochemistry and real-time reverse transcriptase PCR revealed that isoflurane administration inhibited formalin injection-induced c-fos expression in the spinal cord. Formaldehyde 111-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-143 20977938-11 2011 Frontal Fos expression was decreased after lamotrigine and valproate, but not after riboflavin. Lamotrigine 43-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-11 20977938-11 2011 Frontal Fos expression was decreased after lamotrigine and valproate, but not after riboflavin. Valproic Acid 59-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-11 21404708-0 2011 [Effects of polychlorinated biphenyl on the expressions of c-fos, c-Myc and beta-catenin in the rat testis]. Polychlorinated Biphenyls 12-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 21585054-0 2011 [Effects of electroacupuncture on expression of tyrosine hydroxylase and c-fos in hippocampal CA 1 area in ketamine-addiction rats]. Ketamine 107-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 21585054-14 2011 CONCLUSION: EA of "Zusanli"(ST 36)-"Sanyinjiao" (SP 6) can downregulate ketamine-addiction induced increase of expression of tyrosine hydroxylase and c-fos in the hippocampal CA 1 region in ketamine-addiction rats, which may contribute to its effect in relieving ketamine addiction symptoms in clinic. Ketamine 72-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-155 21585054-14 2011 CONCLUSION: EA of "Zusanli"(ST 36)-"Sanyinjiao" (SP 6) can downregulate ketamine-addiction induced increase of expression of tyrosine hydroxylase and c-fos in the hippocampal CA 1 region in ketamine-addiction rats, which may contribute to its effect in relieving ketamine addiction symptoms in clinic. Ketamine 190-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-155 21585054-14 2011 CONCLUSION: EA of "Zusanli"(ST 36)-"Sanyinjiao" (SP 6) can downregulate ketamine-addiction induced increase of expression of tyrosine hydroxylase and c-fos in the hippocampal CA 1 region in ketamine-addiction rats, which may contribute to its effect in relieving ketamine addiction symptoms in clinic. Ketamine 190-198 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-155 21404708-5 2011 The expressions of c-fos and c-Myc were significantly higher in the 1 and 10 mg/kg PCB groups than in the control and 0.1 mg/kg PCB groups (P < 0.01). Polychlorinated Biphenyls 83-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 21404708-5 2011 The expressions of c-fos and c-Myc were significantly higher in the 1 and 10 mg/kg PCB groups than in the control and 0.1 mg/kg PCB groups (P < 0.01). Polychlorinated Biphenyls 128-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 21404708-7 2011 CONCLUSION: PCB causes changes in the phenotype of the testis tissue, and the abnormal expressions of c-fos, c-Myc and beta-catenin are closely related to the PCB-induced testis injury. Polychlorinated Biphenyls 159-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 20851150-0 2011 APM/CD13 and FOS in the hypothalamus of monosodium glutamate obese and food deprived rats. Sodium Glutamate 40-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 20977893-0 2011 Fos expression induced by activation of NMDA and neurokinin-1 receptors in the trigeminal subnucleus caudalis in vitro: role of protein kinases. N-Methylaspartate 40-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 21129389-10 2011 BN82451 treatment significantly reversed the overexpression of c-Fos, FosB and Arc mRNA associated with the dyskinesiogenic action of L-dopa. Levodopa 134-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 21129389-11 2011 A significant correlation between the degree of overexpression of c-Fos, FosB and Arc mRNA and the dyskinesiogenic action of L-dopa was observed. Levodopa 125-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 20977893-8 2011 NMDA and [Sar(9),Met(O(2))(11)]-SP significantly increased Fos expression, but these increases could be prevented by chelerythrine chloride. N-Methylaspartate 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 20977893-8 2011 NMDA and [Sar(9),Met(O(2))(11)]-SP significantly increased Fos expression, but these increases could be prevented by chelerythrine chloride. met(o(2))( 17-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 20977893-8 2011 NMDA and [Sar(9),Met(O(2))(11)]-SP significantly increased Fos expression, but these increases could be prevented by chelerythrine chloride. TFF2 protein, human 29-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 20977893-8 2011 NMDA and [Sar(9),Met(O(2))(11)]-SP significantly increased Fos expression, but these increases could be prevented by chelerythrine chloride. chelerythrine 117-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 20977893-9 2011 Rp-cAMP had no effect on Fos induced by NMDA but caused a significant reduction in Fos induced by [Sar(9),Met(O(2))(11)]-SP. Cyclic AMP 3-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 20971082-8 2011 BDA-labeled fibers from the SCN and vSPVZ formed appositions with orexin neurons and gonadotropin-releasing hormone neurons, two cell populations whose rhythms in Fos expression track temporally reversed patterns of locomotor and reproductive behavior, respectively, in diurnal and nocturnal rodents. biotinylated dextran amine 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 20934408-0 2011 The Fos expression in rat brain following electrical stimulation of dura mater surrounding the superior sagittal sinus changed with the pre-treatment of rizatriptan benzoate. rizatriptan 153-173 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-7 20934408-3 2011 With the pre-treatment of intraperitoneal injection of rizatriptan benzoate, the number of Fos-like immunoreactive neurons decreased in the spinal trigeminal nucleus caudal part and raphe magnus nucleus, increased in the periaqueductal gray, and remained unchanged in the ventromedial hypothalamic nucleus and mediodorsal thalamus nucleus. rizatriptan 55-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-94 20801158-7 2011 Also, 0.5 mug AP-5 prior to conditioning significantly reduced c-fos expression in response to the CS in nucleus accumbens core. 2-amino-5-phosphopentanoic acid 14-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 20801158-7 2011 Also, 0.5 mug AP-5 prior to conditioning significantly reduced c-fos expression in response to the CS in nucleus accumbens core. Cesium 99-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 21117907-8 2011 We also found that SB431542 (transforming growth factor-beta (TGF-beta) receptor kinases inhibitor) suppressed ultrasound-induced MMP?13 and c-Fos gene expression, and p38 phosphorylation. 4-(5-benzo(1,3)dioxol-5-yl-4-pyridin-2-yl-1H-imidazol-2-yl)benzamide 19-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 21056613-8 2011 Results of immunoprecipitation indicated that nuclear c-fos/c-jun heterodimer increases with andrographolide treatment. andrographolide 93-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 21161464-2 2011 In RWIS rats Fos immunoreactive (Fos-IR) nuclei dramatically increased in the paraventricular nucleus (PVN), the supraoptic nucleus (SON), the dorsal motor nucleus of the vagus (DMV), the nucleus of the solitary tract (NTS), the area postrema (AP), and the ventrolateral medulla (VLM). (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 178-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 20832430-4 2011 AJS induced a significant increase in c-Fos-like immunoreactivity (FLI) throughout the caudal-rostral extent of the basolateral, medial, and central (CEA) subnuclei of the amygdala. ajs 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 22312458-3 2011 Following injection of L-proline, the number of activated hypothalamic neurons that coexpressed vasopressin and c-Fos was much greater in the supraoptic nucleus (SON) than in the paraventricular nucleus (PVN) of rats with increased blood pressure. Proline 23-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 21161464-2 2011 In RWIS rats Fos immunoreactive (Fos-IR) nuclei dramatically increased in the paraventricular nucleus (PVN), the supraoptic nucleus (SON), the dorsal motor nucleus of the vagus (DMV), the nucleus of the solitary tract (NTS), the area postrema (AP), and the ventrolateral medulla (VLM). (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 178-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 21161464-5 2011 In RWIS and nonstressed rats, the percentage of Fos-IR nuclei in TH-IR neurons was 38.0 and 14.3% in the DMV, 34.4 and 9.7% in the NTS, 18.6 and 4.5% in the AP, and 45.7 and 18.9% in the VLM, respectively. (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 105-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 21934352-7 2011 The result showed that intrathecal PF preconditioning was effective to suppress spinal c-Fos expression in both superficial (lamina I-II) and deep (lamina IV-VI) layers of the L(4-5) dorsal spine. peoniflorin 35-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 22140566-4 2011 5-OH-HxMF caused the enhancement of cyclic AMP response element binding protein (CREB) phosphorylation, c-fos gene expression and CRE-mediated transcription, which was inhibited by 2-naphthol AS-E phosphate (KG-501), a specific antagonist for the CREB-CBP complex formation. 5-hydroxy-3,6,7,8,3',4'-hexamethoxyflavone 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 22140566-4 2011 5-OH-HxMF caused the enhancement of cyclic AMP response element binding protein (CREB) phosphorylation, c-fos gene expression and CRE-mediated transcription, which was inhibited by 2-naphthol AS-E phosphate (KG-501), a specific antagonist for the CREB-CBP complex formation. 2-naphthol as-e phosphate 181-206 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 21980505-6 2011 Some FG/TMR-DA double-labeled premotor neurons, which were observed bilaterally in the PCRt, MdD, dorsal part of the MdV, peri-motor nucleus regions, contacted with BDA-labeled axonal terminals and expressed c-fos protein-like immunoreactivity which induced by subcutaneous injection of formalin into the lip. tmr 8-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 208-213 20888358-6 2011 This dose of prazosin (1.0 mg/kg) was then administered 30 min prior to VCS or control scapular stimulation (CSS) and Fos-IR was examined in the posterodorsal medial amygdala (MeaPD), the medial preoptic area (mPOA), and the rostral ventromedial hypothalamus (rVMH). Prazosin 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 20888358-8 2011 Prazosin treatment inhibited the increase in Fos-IR in the mPOA and MeaPD but not in the rVMH. Prazosin 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 21980505-6 2011 Some FG/TMR-DA double-labeled premotor neurons, which were observed bilaterally in the PCRt, MdD, dorsal part of the MdV, peri-motor nucleus regions, contacted with BDA-labeled axonal terminals and expressed c-fos protein-like immunoreactivity which induced by subcutaneous injection of formalin into the lip. biotinylated dextran amine 165-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 208-213 21858088-6 2011 In the RWIS rats, Fos-IR neurons were identified in 31% of OT-IR neurons and 40% of AVP-IR neurons in the PVN, while in the SON it represented 28%, 53% respectively; (3) V(1b)R-IR and OTR-IR neurons occupied all portions of the NTS and DMV. (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 236-239 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 21957454-7 2011 Additional labeling with c-Fos was seen in the subnucleus interpolaris of the spinal trigeminal nucleus, the rostral ventrolateral medulla, the superior salivatory nucleus, the rostral ventromedial medulla, and the A1, A5, A7 and subcoeruleus catecholamine areas. Catecholamines 243-256 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 21179447-9 2010 METH administration caused increases in the expression of c-fos, Egr1, and Nor-1 on the intact side. Methamphetamine 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 21887361-9 2011 Hypoxia-inducible factor 1alpha (HIF-1alpha) and phosphorylated mitogen-activated protein kinase (pMAPK) were considered as possible mediators of IH-induced changes in neurogenesis and c-Fos expression. Ile-His 146-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 185-190 21525724-0 2011 Effect of long-term prazosin and yohimbine administration on c-Fos expression in spinal neurons: inhibitory effect of alpha-1 and alpha-2 adrenoceptors on afferents from the lower urinary tract. Prazosin 20-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 21525724-0 2011 Effect of long-term prazosin and yohimbine administration on c-Fos expression in spinal neurons: inhibitory effect of alpha-1 and alpha-2 adrenoceptors on afferents from the lower urinary tract. Yohimbine 33-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 21525724-6 2011 The numbers of c-Fos-positive neurons in the dorsal horn were significantly lower in rats that received prazosin than in controls. Prazosin 104-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 21525724-7 2011 Yohimbine reduced the number of c-Fos-positive neurons in part of the dorsal horn. Yohimbine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 20486760-0 2010 Reactive Oxygen Species-Dependent c-Fos/Activator Protein 1 Induction Upregulates Heme Oxygenase-1 Expression by Bradykinin in Brain Astrocytes. Reactive Oxygen Species 0-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 20486760-8 2010 Taken together, these results suggested that in RBA-1 cells, activation of ERK/NF-kappaB and JNK/c-Jun cascades by a Nox/ROS-dependent event enhancing c-Fos/AP-1 activity is essential for HO-1 upregulation and activation induced by BK. Reactive Oxygen Species 121-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-156 21179447-12 2010 These include c-fos and Nor-1 which show greater changes on the normal DA side. Dopamine 71-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 21253331-4 2010 In the present study, we investigated the effects of vardenafil on oxytocin and c-Fos expression in the paraventricular nucleus (PVN) of conscious rats. Vardenafil Dihydrochloride 53-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 20570698-0 2010 Patterns of social-experience-related c-fos and Arc expression in the frontal cortices of rats exposed to saccharin or moderate levels of ethanol during prenatal brain development. Ethanol 138-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 20570698-2 2010 Fetal-ethanol-related reductions in the expression of the immediate early genes (IEGs) c-fos and Arc and alterations in dendritic spine density in ventrolateral and medial aspects of frontal cortex suggest a dissociation reminiscent of that described by Kolb et al. Ethanol 6-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 20883707-2 2010 To clarify the mechanisms of chemotherapy-induced anorexia, we studied the expression of c-fos and appetite-regulating neuropeptidergic and inflammatory mediators in the hypothalamus of rats treated with methotrexate (MTX). Methotrexate 204-216 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 20883707-10 2010 CONCLUSION: The pattern of c-fos expression in the hypothalamus during MTX treatment is similar to that seen with systemic dehydration, which is known to cause anorexia. Methotrexate 71-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 20926762-10 2010 Moreover, benzamil treatment decreased c-Fos immunoreactivity in the SON and in medial parvocellular and posterior magnocellular neurons of the PVN in DOCA-salt rats but not areas associated with regulation of sympathetic activity. benzamil 10-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 20637816-6 2010 Fos immunoreactivity in brain areas associated with sodium appetite and excretion were not influenced by diet. Sodium 52-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 21253331-11 2010 c-Fos in the oxytocin neurons of the PVN was increased by 1 day administration of 2 mg/kg vardenafil, and this effect of vardenafil also appeared in a duration-dependent manner. Vardenafil Dihydrochloride 90-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 21253331-11 2010 c-Fos in the oxytocin neurons of the PVN was increased by 1 day administration of 2 mg/kg vardenafil, and this effect of vardenafil also appeared in a duration-dependent manner. Vardenafil Dihydrochloride 121-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 20847126-8 2010 Furthermore, a 5-HT(2A) antagonist (ketanserin) significantly reduced 3-G (120 min) load-induced Fos expression in the medial vestibular nucleus, and chronically administered ketanserin ameliorated hypergravity-induced hypophagia. Ketanserin 36-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 20727386-6 2010 Recently, increased c-fos expression by systemic PCP administration was reported in the mediodorsal nucleus of the thalamus (MD) and the centromedial nucleus of the thalamus (CM), which have strong reciprocal innervations with the mPFC. Phencyclidine 49-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 20849934-1 2010 The brain"s response to ethanol intake has been extensively investigated using electrophysiological recordings, brain lesion techniques, and c-Fos immunoreactivity. Ethanol 24-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 20844266-5 2010 Water deprivation increased c-Fos staining in the lamina terminalis. Water 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 20678554-5 2010 Intraperitoneal administration of an inhibitor for COX-1, COX-2 or inducible NOS (iNOS), but not for neuronal NOS (nNOS), reduced RS-induced elevation of plasma catecholamine levels and Fos expression in the presympathetic PVN neurons. rs 130-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 186-189 20731630-8 2010 Strikingly, oxytocin significantly reduced methamphetamine-induced Fos expression in two regions of the basal ganglia: the subthalamic nucleus and the nucleus accumbens core. Methamphetamine 43-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 20965221-7 2011 In addition, water deprivation induced more c-fos expression in the central dipsogenic areas, including the paraventricular and supraoptic nuclei in the offspring exposed to prenatal high sucrose. Water 13-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 20965221-7 2011 In addition, water deprivation induced more c-fos expression in the central dipsogenic areas, including the paraventricular and supraoptic nuclei in the offspring exposed to prenatal high sucrose. Sucrose 188-195 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 20816675-5 2010 Both pontine cell groups express the transcription factor FoxP2 and become c-Fos activated following sodium depletion. Sodium 101-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 21328975-9 2010 The neuroprotection of curcumine in SHR is related to c-jun and c-fos. Curcumin 23-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 20643195-2 2010 The aim of this study was to use c-fos like immunohistochemistry (Fos-LI) to map the trigeminal brainstem complex after intravitreal microinjection or ocular surface application of capsaicin, a selective transient receptor potential vanilloid 1 (TRPV1) agonist in male rats under barbiturate anesthesia. Capsaicin 181-190 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 20643195-2 2010 The aim of this study was to use c-fos like immunohistochemistry (Fos-LI) to map the trigeminal brainstem complex after intravitreal microinjection or ocular surface application of capsaicin, a selective transient receptor potential vanilloid 1 (TRPV1) agonist in male rats under barbiturate anesthesia. Capsaicin 181-190 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 20643195-2 2010 The aim of this study was to use c-fos like immunohistochemistry (Fos-LI) to map the trigeminal brainstem complex after intravitreal microinjection or ocular surface application of capsaicin, a selective transient receptor potential vanilloid 1 (TRPV1) agonist in male rats under barbiturate anesthesia. barbituric acid 280-291 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 20643195-4 2010 In non-inflamed controls, intravitreal capsaicin produced peaks of Fos-LI at the trigeminal subnucleus interpolaris/caudalis (Vi/Vcvl) transition and in superficial laminae at the caudalis/upper cervical cord (Vc/C1) junction regions. Capsaicin 39-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 20643195-5 2010 At the Vc/C1 junction intravitreal capsaicin induced Fos-LI in a dose-dependent manner, while at the Vi/Vcvl transition responses were similar after vehicle or capsaicin injections. Capsaicin 35-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 20643195-6 2010 Two days, but not 7 days, after UV irradiation intravitreal and ocular surface capsaicin-evoked Fos-LI at the Vc/C1 junction and nucleus tractus solitarius (NTS) were markedly enhanced, whereas the responses at the Vi/Vcvl transition were not different from non-inflamed controls. Capsaicin 79-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-99 20554014-8 2010 Intrathecal injection of BBG suppressed the up-regulation of microglial P2X7Rs and increased expression of Fos in the spinal superficial dorsal horn. bbg 25-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 21287731-1 2010 OBJECTIVE: To explore the changes of c-fos in apoptosis of cerebellar granular neuron of neonatal SD rats induced by heroin and the mechanisms of neuronal injury caused by heroin. Heroin 117-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 20600661-1 2010 We previously reported that treatment of prepubertal male rats with low, injected or oral, doses of methylphenidate stimulated cfos, fosB and arc expression in many areas of the developing brain. Methylphenidate 100-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 127-131 21287731-4 2010 RESULTS: Ten microg/mL of heroin was the optimal dose to induce the apoptosis of cerebellar granular neuron at 48 h. Both Western blotting and RT-PCR showed down regulation of c-fos expression. Heroin 26-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 176-181 21287731-5 2010 CONCLUSION: Heroin could induce apoptosis of cerebellar granular neuron and down regulation of c-fos, which may be one of the apoptosis mechanisms. Heroin 12-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 20633205-6 2010 Interestingly, tolerance to cocaine-induced cFos induction was dependent on volitional control of cocaine intake in ventral but not dorsal striatal regions, whereas regulation of FosB and DeltaFosB was similar in cocaine self-administering and yoked animals. Cocaine 28-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-48 20633205-6 2010 Interestingly, tolerance to cocaine-induced cFos induction was dependent on volitional control of cocaine intake in ventral but not dorsal striatal regions, whereas regulation of FosB and DeltaFosB was similar in cocaine self-administering and yoked animals. Cocaine 98-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-48 20600661-3 2010 We report here, for the first time, that low threshold doses of oral ADD, an extended-release mixture of amphetamine salts, now routinely used for the treatment of Attention Deficit Hyperactivity Disorder (ADHD), also increased cfos expression in infantile (postnatal day 10; PD10) and prepubertal (PD24) rat brain. Amphetamine 105-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 228-232 20497419-14 2010 Following a series of micturition reflexes elicited by infusion of saline into the bladder, the immediate early gene product c-Fos was observed in neurons of the lumbosacral IML and approximately 20% of these also expressed ghrelin receptor gene transcripts. Sodium Chloride 67-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 125-130 20680358-0 2010 Repeated administration of methamphetamine blocked cholecystokinin-octapeptide injection-induced c-fos mRNA expression without change in capsaicin-induced junD mRNA expression in rat cerebellum. Methamphetamine 27-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 20335078-2 2010 The purpose of this study is to examine the correlation between pharmacologically-induced changes in cystometric parameters and spinal c-Fos expression in anesthetized rats with bladder hyperactivity induced by the intravesical infusion of acetic acid. Acetic Acid 240-251 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 20716371-0 2010 Administration of the GABAA receptor antagonist picrotoxin into rat supramammillary nucleus induces c-Fos in reward-related brain structures. Picrotoxin 48-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 20677375-9 2010 Adolescent rats with 6-OHDA lesions subjected to restraint stress had greater c-fos expression in the AcbC, AcbSh, DG, Ce, BL, and Tc, compared to the sham and control groups, whereas these differences were not observed among the adult groups. Oxidopamine 21-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 20677375-9 2010 Adolescent rats with 6-OHDA lesions subjected to restraint stress had greater c-fos expression in the AcbC, AcbSh, DG, Ce, BL, and Tc, compared to the sham and control groups, whereas these differences were not observed among the adult groups. 1-aminocyclobutanecarboxylic acid 102-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 20677375-9 2010 Adolescent rats with 6-OHDA lesions subjected to restraint stress had greater c-fos expression in the AcbC, AcbSh, DG, Ce, BL, and Tc, compared to the sham and control groups, whereas these differences were not observed among the adult groups. acbsh 108-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 20677375-9 2010 Adolescent rats with 6-OHDA lesions subjected to restraint stress had greater c-fos expression in the AcbC, AcbSh, DG, Ce, BL, and Tc, compared to the sham and control groups, whereas these differences were not observed among the adult groups. 4-Butyrolactone 123-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 20553875-8 2010 Chronic estradiol treatment modulated the neural response in some regions: less c-Fos was induced in the superior vestibular nucleus and locus coeruleus after estradiol replacement; estradiol treatment eliminated the asymmetry of c-Fos expression in the locus coeruleus and supragenualis nucleus, created an asymmetry in the prepositus nucleus and reversed the asymmetry in the parabrachial nucleus. Estradiol 8-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 20553875-8 2010 Chronic estradiol treatment modulated the neural response in some regions: less c-Fos was induced in the superior vestibular nucleus and locus coeruleus after estradiol replacement; estradiol treatment eliminated the asymmetry of c-Fos expression in the locus coeruleus and supragenualis nucleus, created an asymmetry in the prepositus nucleus and reversed the asymmetry in the parabrachial nucleus. Estradiol 8-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 230-235 20570662-4 2010 Following EIS, tonotopic c-Fos expression was observed for each stimulation time in ipsilateral AVCN, DCN bilaterally, and contralateral CIC. dcn 102-105 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 20570662-9 2010 In DCN, the density of large c-Fos positive nuclei fell in the upper and rose in the deep layers from 45 min to 5h of EIS. dcn 3-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 20427724-7 2010 Injection of LiCl + antagonist compared with LiCl alone generated an increase in c-Fos immunoreactivity in the central nucleus of the amygdala. Lithium Chloride 13-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 20427724-7 2010 Injection of LiCl + antagonist compared with LiCl alone generated an increase in c-Fos immunoreactivity in the central nucleus of the amygdala. Lithium Chloride 45-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 20056558-4 2010 At the PAG, increases in Fos expression were detected until the 4th week, with a reversal to baseline values at 10 weeks in all areas except the ventrolateral PAG. pag 7-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 20056558-4 2010 At the PAG, increases in Fos expression were detected until the 4th week, with a reversal to baseline values at 10 weeks in all areas except the ventrolateral PAG. pag 159-162 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 20056558-5 2010 Co-localisation of Fos with NeuN ascertained the neuronal nature of Fos-expressing cells at the spinal cord and PAG. pag 112-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 20056558-8 2010 Gabapentin decreased Fos expression at the spinal cord and PAG and reversed mechanical hypersensitivity. Gabapentin 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 20400272-0 2010 Increased voluntary ethanol consumption and c-Fos expression in selected brain areas induced by fear memory retrieval in ethanol withdrawn rats. Ethanol 121-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 20400272-8 2010 In ethanol withdrawn rats, context-dependent memory retrieval was accompanied by an increased c-Fos expression in the basolateral amygdala, ventrolateral periaqueductal gray, dentate gyrus and dorsomedial periaqueductal gray. Ethanol 3-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 20713560-5 2010 Results revealed that acute paracetamol administration substantially decreased the number of Fos-immunoreactive cells in the parietal cortex and TNC without causing change in CSD frequency. Acetaminophen 28-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 20713560-6 2010 On the other hand, chronic paracetamol administration led to an increase in CSD frequency as well as CSD-evoked Fos expression in parietal cortex and TNC, indicating an increase in cortical excitability and facilitation of trigeminal nociception. Acetaminophen 27-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 20685882-7 2010 In a second series of experiments, the effect of SB-334867-A and SNAP-7941 alone or combination on the expression of the Fos protein was determined. Antimony 49-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-124 20680358-9 2010 Thus, we conclude that repeated methamphetamine administration though dopamine selectively inhibits the c-fos mRNA expression after CCK-8 injection in the cerebellum. Methamphetamine 32-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 20680358-9 2010 Thus, we conclude that repeated methamphetamine administration though dopamine selectively inhibits the c-fos mRNA expression after CCK-8 injection in the cerebellum. Dopamine 70-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 20797570-9 2010 In the uranyl acetate-induced ARF, activation of Fos and Fra-2 immunoreactivity took place at the earliest time-point (3 hours) which persisted even after improvement of ARF. uranyl acetate 7-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-52 20623106-10 2010 QNP-reduced BDNF expression in the hippocampus and increased c-Fos in the prefrontal cortex. Quinpirole 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 20844807-10 2010 In addition, C. sinensis extract also significantly inhibited the expression of PCNA, c-jun, and c-fos in these PASMCs. pasmcs 112-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 20680358-5 2010 Repeated administration of methamphetamine suppressed the CCK-8-induced c-fos mRNA expression in the rat cerebellum. Methamphetamine 27-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 20663035-0 2010 Effect of gamma-glutamylcysteine ethylester on the levels of c-fos mRNA expression, glutathione and reactive oxygen species formation in kainic acid excitotoxicity. gamma-glutamylcysteine 10-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 20663035-1 2010 OBJECTIVES: The aim of this study was to investigate the effect of gamma-glutamylcysteine ethylester (GCEE), a precursor of glutathione biosynthesis, on the levels of glutathione, formation of reactive oxygen species and c-fos mRNA expression in rat hippocampus and cortex in kainic acid-induced excitotoxicity. gamma-glutamylcysteine ethylester 67-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 221-226 20663035-1 2010 OBJECTIVES: The aim of this study was to investigate the effect of gamma-glutamylcysteine ethylester (GCEE), a precursor of glutathione biosynthesis, on the levels of glutathione, formation of reactive oxygen species and c-fos mRNA expression in rat hippocampus and cortex in kainic acid-induced excitotoxicity. gcee 102-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 221-226 20663035-6 2010 KEY FINDINGS: Kainic acid treatment significiantly upregulated the expression of c-fos mRNA in the hippocampus and cortex when compared to the control group. Kainic Acid 14-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 20434770-11 2010 Lastly, alarin stimulated Fos induction in hypothalamic nuclei, such as the paraventricular nucleus and the nucleus of the tractus solitarious. alarin 8-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 20813664-4 2010 RESULTS: The rats with acute sodium depletion exhibited significantly more numerous c-fos-positive neurons and GABA/Fos double-labeled neurons in the CeA than the control group (P<0.01, P<0.05). Sodium 29-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 20813664-4 2010 RESULTS: The rats with acute sodium depletion exhibited significantly more numerous c-fos-positive neurons and GABA/Fos double-labeled neurons in the CeA than the control group (P<0.01, P<0.05). Sodium 29-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 20813664-5 2010 Consumption of 0.3 mol/L NaCl significantly increased the number of c-fos and GABA/Fos double labeled neurons compared to the distilled water group (P<0.001, P<0.01). Sodium Chloride 25-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 20813664-5 2010 Consumption of 0.3 mol/L NaCl significantly increased the number of c-fos and GABA/Fos double labeled neurons compared to the distilled water group (P<0.001, P<0.01). Sodium Chloride 25-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 20149549-7 2010 In the CNS, mifepristone increased c-Fos expression in all subdivisions of the medial prefrontal cortex (mPFC) and decreased neuronal activity in some subdivisions of the hippocampus including the CA2, CA3, and hilus region of the dentate gyrus in animals exposed to FST. Mifepristone 12-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 20670470-0 2010 Altered Fos immunoreactivity in the hypothalamus after glucose administration in pre- and post-weaning malnourished rats. Glucose 55-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-11 20149549-8 2010 In contrast, mifepristone increased neuronal activity in the ventral subiculum (output region of the hippocampus) and decreased c-Fos expression in the central amygdala (CeA) in animals exposed to FST. Mifepristone 13-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-133 20214928-12 2010 The c-fos and Hsp70 mRNA expressions were positively correlated to the highest SA levels occurring 45 min before sacrifice, suggesting that high SA and frontal cortex activation are related. sa 79-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 20214928-12 2010 The c-fos and Hsp70 mRNA expressions were positively correlated to the highest SA levels occurring 45 min before sacrifice, suggesting that high SA and frontal cortex activation are related. sa 145-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 20637119-0 2010 Propofol induces MAPK/ERK cascade dependant expression of cFos and Egr-1 in rat hippocampal slices. Propofol 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-62 20637119-3 2010 The underlying mechanism for this memory loss is unclear but may potentially be related to the induction of memory-associated genes such as c-Fos and Egr-1 by propofol. Propofol 159-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-145 20637119-4 2010 This study explored the effects of propofol on c-Fos and Egr-1 expression in rat hippocampal slices. Propofol 35-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 20637119-8 2010 We demonstrate that propofol induced the expression of c-Fos and Egr-1 within 30 and 60 min of exposure time. Propofol 20-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 20637119-9 2010 At 16.8 muM concentration, propofol induced a 110% increase in c-Fos transcription and 90% decrease in the transcription of Egr-1. Propofol 27-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 20637119-11 2010 Propofol-induced c-Fos and Egr-1 transcription was abolished by inhibitors of RAS, RAF, MEK, ERK and p38-MAPK in the MAPK/ERK cascade. Propofol 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 20637119-12 2010 CONCLUSIONS: Our study shows that clinically relevant concentrations of propofol induce c-Fos and down regulated Egr-1 expression via an MAPK/ERK mediated pathway. Propofol 72-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 20637119-13 2010 We demonstrated that propofol induces a time and dose dependant transcription of IEGs c-Fos and Egr-1 in rat hippocampal slices. Propofol 21-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 20399242-8 2010 The percentage of c-Fos-positive oxytocin cells in the hypothalamic paraventricular nucleus was significantly lower in rats chronically exposed to the HS than to the CS diet, regardless of which diet they received on the final day. Starch 166-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 20514481-5 2010 Ultrasound-induced c-Fos expression was measured in different periaqueductal gray (PAG) and amygdala subregions after treatment with diazepam or Ro 64-6198. Diazepam 133-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 20514481-7 2010 Ultrasound application increased c-Fos expression in the dorsal and dorsolateral PAG (dPAG, dlPAG) and amygdaloid subregions. dpag 86-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 20514481-7 2010 Ultrasound application increased c-Fos expression in the dorsal and dorsolateral PAG (dPAG, dlPAG) and amygdaloid subregions. dlpag 92-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 20514481-8 2010 Diazepam, but not Ro 64-6198, reduced c-Fos expression in the dPAG/dlPAG, while Ro 64-6198, but not diazepam, reduced c-Fos expression in the central amygdala. Diazepam 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-43 20406670-0 2010 The brain pattern of c-fos induction by two doses of amphetamine suggests different brain processing pathways and minor contribution of behavioural traits. Amphetamine 53-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 20406670-3 2010 In the present experiment, we studied in adult male rats the c-fos expression induced by two doses of AMPH (1.5 and 5 mg/kg sc) in a wide range of brain areas, and investigated the possible contribution of novelty-induced activity and anxiety traits. Amphetamine 102-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 20406670-4 2010 AMPH administration increased Fos+ neurons in an important number of telencephalic, diencephalic and brainstem areas. Amphetamine 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 20605533-8 2010 Western Blot analysis confirmed that CHX potently inhibited PTZ-induced protein synthesis (c-Fos) in the rat brain, examined 60min after CHX and PTZ administration. Cycloheximide 37-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 20605533-8 2010 Western Blot analysis confirmed that CHX potently inhibited PTZ-induced protein synthesis (c-Fos) in the rat brain, examined 60min after CHX and PTZ administration. Pentylenetetrazole 60-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 20375114-2 2010 Among the CREB target genes, some immediate early genes exist that are responsive to cAMP including the nur77 and c-fos genes. Cyclic AMP 85-89 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 20739787-7 2010 The effect of flunarizine on c-fos expression and its efficacy in restoring neural function was evaluated. Flunarizine 14-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 20739787-8 2010 RESULTS: The c-fos messenger ribonucleic acid (mRNA) and protein expression in FI and FII groups was significantly lower than in the saline group and was the least in the FII group. Sodium Chloride 133-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 20739787-11 2010 CONCLUSIONS: When administered after crush injury to peripheral nerves, flunarizine may protect neurons with lesions from further damage and improve neural function by downregulating c-fos expression. Flunarizine 72-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 183-188 20677375-0 2010 Age-specific effects of 6-hydroxydopamine lesions of the rat medial prefrontal cortex on stress-induced c-fos expression in subcortical areas. Oxidopamine 24-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 20677375-2 2010 Therefore, we investigated the effects of 6-OHDA lesions of the mPFC in adolescent and adult rats on stress-induced c-fos expression in the brain. Oxidopamine 42-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-121 20463049-9 2010 The similar pattern of Fos expression in catecholamine cell groups after GH and ghrelin and the prolonged GH secretion in response to ghrelin in DSAP rats together suggest that activation of hindbrain catecholamine neurons by ghrelin or GH could be a component of a negative feedback response controlling GH levels. Catecholamines 41-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 20463049-9 2010 The similar pattern of Fos expression in catecholamine cell groups after GH and ghrelin and the prolonged GH secretion in response to ghrelin in DSAP rats together suggest that activation of hindbrain catecholamine neurons by ghrelin or GH could be a component of a negative feedback response controlling GH levels. Ghrelin 80-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 20127074-8 2010 The transcription factor c-fos was up-regulated in hippocampus, and c-jun was elevated both in cortex and hippocampus in PFOS-treated groups. perfluorooctane sulfonic acid 121-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 20338222-4 2010 To determine whether these nonfornical pathways might mediate forebrain activation after VH NMDA infusion, we examined the effects of bilateral VH NMDA infusion on c-Fos protein expression in the mPFC and nucleus accumbens (NAC) after sham vs. bilateral fornix lesions. N-Methylaspartate 147-151 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-169 20338222-5 2010 Significant increases of c-Fos expression were observed in both the mPFC and NAC after bilateral VH NMDA infusions. N-Methylaspartate 100-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 20374215-14 2010 Ethanol administration caused a significant decrease in the number of BF wake-promoting neurons with c-Fos immunoreactivity. Ethanol 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 101-106 20608965-7 2010 Double-labelling in the LS of IMO-exposed rats revealed that arc was expressed in only one-third of Fos+ neurons, suggesting two populations of Fos+ neurons. 6-o-phosphoryl inosine monophosphate 30-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 144-147 20210978-4 2010 METHODS: Three daily enemas containing 2,4,6-trinitrobenzene sulfonic acid (TNBS), which were given to rats produced chronic colitis and ongoing activation of Fos in brain neurons. Trinitrobenzenesulfonic Acid 39-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-162 20210978-4 2010 METHODS: Three daily enemas containing 2,4,6-trinitrobenzene sulfonic acid (TNBS), which were given to rats produced chronic colitis and ongoing activation of Fos in brain neurons. Trinitrobenzenesulfonic Acid 76-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-162 20406698-5 2010 In addition, the increase in malondialdehyde content and the up-regulation of c-fos mRNA expression of the slices induced by hypoxia was significantly reduced by NaHS. sodium bisulfide 162-166 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 20406698-6 2010 These results indicate that exogenous H(2)S may protect the medullary respiratory center against hypoxic injury via activation of K(ATP) channels, reduction of lipid peroxidation and down-regulation of c-fos. Hydrogen Sulfide 38-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 202-207 20156467-7 2010 Immunohistochemical analysis found that the brain response to formalin injection was also more sensitive in middle-aged rats than young rats; a significant increase in the number of c-Fos immunoreactive cells was found in the ventral portion of the lateral septum of middle-aged rats injected with formalin compared to young and middle-aged rats injected with saline, irrespective of estrous cyclicity. Formaldehyde 62-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 182-187 20156467-7 2010 Immunohistochemical analysis found that the brain response to formalin injection was also more sensitive in middle-aged rats than young rats; a significant increase in the number of c-Fos immunoreactive cells was found in the ventral portion of the lateral septum of middle-aged rats injected with formalin compared to young and middle-aged rats injected with saline, irrespective of estrous cyclicity. Formaldehyde 298-306 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 182-187 20156467-7 2010 Immunohistochemical analysis found that the brain response to formalin injection was also more sensitive in middle-aged rats than young rats; a significant increase in the number of c-Fos immunoreactive cells was found in the ventral portion of the lateral septum of middle-aged rats injected with formalin compared to young and middle-aged rats injected with saline, irrespective of estrous cyclicity. Sodium Chloride 360-366 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 182-187 20159026-7 2010 In all three rearing groups, Fos activation within brainstem and forebrain regions of interest was significantly enhanced after LiCl vs. saline. Lithium Chloride 128-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 20159026-7 2010 In all three rearing groups, Fos activation within brainstem and forebrain regions of interest was significantly enhanced after LiCl vs. saline. Sodium Chloride 137-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 20302915-4 2010 Rats receiving 1.5-g/kg ethanol showed an increase in Fos-ir in the MPOA. Ethanol 24-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 20200127-6 2010 The chronic intermittent sucrose consumption significantly lowered, in the SIA and SIR rats, the levels of expression of corticotropin-releasing factor type 2 receptor and restraint stress-induced expression of c-fos mRNA in the medioventral part of the lateral septum. Sucrose 25-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 211-216 19786358-5 2010 Formalin-evoked nociceptive behaviour was associated with increased Fos immunoreactivity (FI) in the CA2/3 region of the hippocampus and rostral ventromedial medulla, effects attenuated by intra-BLA rimonabant. Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 20228277-5 2010 CO(2) laser irradiation to the gingiva just after tooth movement caused a significant decrease of Fos-IR neurons. co(2) 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 19938175-4 2010 It was found that interruption of astrocyte metabolism with fluorocitrate significantly reduced Fos protein expression in both astrocytes and neurons, whereas blockage of gap junctions with carbenoxolone clearly reduced Fos protein expression in neurons, but not in astrocytes. fluorocitrate 60-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-99 19938175-4 2010 It was found that interruption of astrocyte metabolism with fluorocitrate significantly reduced Fos protein expression in both astrocytes and neurons, whereas blockage of gap junctions with carbenoxolone clearly reduced Fos protein expression in neurons, but not in astrocytes. Carbenoxolone 190-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 220-223 20236223-2 2010 We have previously shown that acute systemic nicotine treatment induces Fos expression in the lateral hypothalamus and perifornical area (LH/PFA), with orexin/hypocretin neurons being particularly responsive. Nicotine 45-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 20236223-8 2010 Lesioned animals showed reduced Fos-positive orexin neurons following nicotine treatment. Nicotine 70-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 20045056-4 2010 We compared the effects of neboglamine, D-serine, clozapine, and haloperidol on the expression of Fos-like immunoreactivity (FLI), a marker of neuronal activation, in rat forebrain. neboglamine 27-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 20045056-4 2010 We compared the effects of neboglamine, D-serine, clozapine, and haloperidol on the expression of Fos-like immunoreactivity (FLI), a marker of neuronal activation, in rat forebrain. D-serine 40-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 20045056-4 2010 We compared the effects of neboglamine, D-serine, clozapine, and haloperidol on the expression of Fos-like immunoreactivity (FLI), a marker of neuronal activation, in rat forebrain. Clozapine 50-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 20045056-4 2010 We compared the effects of neboglamine, D-serine, clozapine, and haloperidol on the expression of Fos-like immunoreactivity (FLI), a marker of neuronal activation, in rat forebrain. Haloperidol 65-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 20109537-6 2010 Second, we determined the effects of CPA and that of an A(1) receptor antagonist, 1,3-dipropyl-8-phenylxanthine (CPDX) into the PF-LHA on cFos-protein immunoreactivity (Fos-IR) in HCRT and non-HCRT neurons around the microdialysis probe used for their delivery. 1,3-dipropyl-8-phenylxanthine 82-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 20109537-6 2010 Second, we determined the effects of CPA and that of an A(1) receptor antagonist, 1,3-dipropyl-8-phenylxanthine (CPDX) into the PF-LHA on cFos-protein immunoreactivity (Fos-IR) in HCRT and non-HCRT neurons around the microdialysis probe used for their delivery. cefpodoxime 113-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 20109537-9 2010 Doses of CPA (50 muM) and CPDX (50 muM) that suppressed and induced arousal, respectively, in our earlier study [Alam MN, Kumar S, Rai S, Methippara M, Szymusiak R, McGinty D (2009) Brain Res 1304:96-104], significantly suppressed and increased Fos-IR in HCRT and non-HCRT neurons. N(6)-cyclopentyladenosine 9-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 245-248 20109537-9 2010 Doses of CPA (50 muM) and CPDX (50 muM) that suppressed and induced arousal, respectively, in our earlier study [Alam MN, Kumar S, Rai S, Methippara M, Szymusiak R, McGinty D (2009) Brain Res 1304:96-104], significantly suppressed and increased Fos-IR in HCRT and non-HCRT neurons. cefpodoxime 26-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 245-248 20236391-0 2010 Functional magnetic resonance imaging and c-Fos mapping in rats following a glucoprivic dose of 2-deoxy-D-glucose. Deoxyglucose 96-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 20631887-7 2010 MCAO markedly increased the expression of c-Fos protein in the insular cortex and cingulate cortex ipsilateral to the occlusion 2 h after MCAO, and pretreatment with MK-801 produced marked reduction of c-Fos protein expression compared to MCAO group (p<0.01). Dizocilpine Maleate 166-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 20631887-7 2010 MCAO markedly increased the expression of c-Fos protein in the insular cortex and cingulate cortex ipsilateral to the occlusion 2 h after MCAO, and pretreatment with MK-801 produced marked reduction of c-Fos protein expression compared to MCAO group (p<0.01). Dizocilpine Maleate 166-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 202-207 20214940-7 2010 In the present study, we revealed that icv administered relaxin-3 induced dense Fos-like immunoreactivity (Fos-LI) in the rat hypothalamus and circumventricular organs including the organum vasculosum of the lamina terminalis, the median preoptic nucleus, supraoptic nucleus (SON), the subfornical organ (SFO) and the paraventricular nucleus (PVN), that are related to the central regulation of body fluid balance. icv 39-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 20214940-7 2010 In the present study, we revealed that icv administered relaxin-3 induced dense Fos-like immunoreactivity (Fos-LI) in the rat hypothalamus and circumventricular organs including the organum vasculosum of the lamina terminalis, the median preoptic nucleus, supraoptic nucleus (SON), the subfornical organ (SFO) and the paraventricular nucleus (PVN), that are related to the central regulation of body fluid balance. icv 39-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 20298714-1 2010 Amphetamines induce stereotypy, which correlates with patch-enhanced c-Fos expression the patch compartment of caudate putamen (CPu). Amphetamines 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-74 20307612-7 2010 After treatment with elevated K(+) and veratridine, c-Fos immunoreactivity appeared in orexin-immunoreactive neurons but not in melanin-concentrating hormone-immunoreactive neurons, suggesting selective excitation of orexin neurons. Veratridine 39-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 20210847-7 2010 Blockade of NK(1) and NK(2) receptors attenuated the formalin-induced increases in mean arterial pressure and heart rate, adrenaline and ACTH concentrations in plasma, and completely abolished the pain-induced c-Fos expression in corticotrophin-releasing hormone neurones localised in the parvocellular division of the PVN. Formaldehyde 53-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 210-215 20233652-6 2010 Exposure to n-3 fatty acids enhances synaptic plasticity by increasing long-term potentiation and synaptic protein expression to increase the dendritic spine density, number of c-Fos-positive neurons and neurogenesis in the hippocampus for learning memory processing. Fatty Acids, Omega-3 12-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 177-182 20132864-0 2010 Peripheral inhibition of glutaminase reduces carrageenan-induced Fos expression in the superficial dorsal horn of the rat. Carrageenan 45-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 20096336-9 2010 Intra-accumbens injections of eticlopride also increased local expression of c-Fos immunoreactivity, and this effect was attenuated by co-administration of MSX-3. eticlopride 30-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 20096751-0 2010 c-Fos identification of neuroanatomical sites associated with haloperidol and clozapine disruption of maternal behavior in the rat. Haloperidol 62-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 20096751-0 2010 c-Fos identification of neuroanatomical sites associated with haloperidol and clozapine disruption of maternal behavior in the rat. Clozapine 78-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 20096751-4 2010 The present study used c-Fos immunohistochemistry technique, together with pharmacological tools and behavioral observations, and delineated the neuroanatomical bases of the disruptive effects of haloperidol and clozapine. Haloperidol 196-207 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 20096751-4 2010 The present study used c-Fos immunohistochemistry technique, together with pharmacological tools and behavioral observations, and delineated the neuroanatomical bases of the disruptive effects of haloperidol and clozapine. Clozapine 212-221 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 20096751-11 2010 Neuroanatomically, the nucleus accumbens (both the shell and core), dorsolateral striatum and lateral septum showed increased c-Fos expression to the treatment of haloperidol. Haloperidol 163-174 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 20096751-12 2010 In contrast, the nucleus accumbens shell showed increased expression of c-Fos to the treatment of clozapine. Clozapine 98-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-77 19816767-1 2010 The purpose of this study is to investigate the change of phosphorylated p44/42 extracellular signal-regulated kinase (pERK1/2) and c-fos expression induced by single-prolonged stress (SPS) in medial prefrontal cortex (mPFC), and to determine whether extracellular signal-regulated kinase (ERK) pathway plays a role in SPS. Sodium phenolsulfonate 185-188 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 19816767-1 2010 The purpose of this study is to investigate the change of phosphorylated p44/42 extracellular signal-regulated kinase (pERK1/2) and c-fos expression induced by single-prolonged stress (SPS) in medial prefrontal cortex (mPFC), and to determine whether extracellular signal-regulated kinase (ERK) pathway plays a role in SPS. Sodium phenolsulfonate 319-322 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 19816767-10 2010 The results of RT-PCR showed that the expression of c-fos mRNA had a significant increase in SPS rats compared with control rats, and the increase was significantly abolished by PD98059. Sodium phenolsulfonate 93-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 20357113-7 2010 AcbSh inactivation increased c-Fos expression in hypothalamus, as well as in paraventricular thalamus and amygdala. acbsh 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 20136691-11 2010 ChIP assays revealed phospho-CREB, c-Jun, Sp1, c-Fos and GR antibodies bound the TRH promoter of cells treated with cAMP or glucocorticoids; anti:RNA-polymerase II immunoprecipitated TRH promoter in a similar proportion as anti:pCREB or anti:GR. Cyclic AMP 116-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 20132864-6 2010 In this study, we measured the effects of glutaminase inhibition on carrageenan-induced spinal Fos expression. Carrageenan 68-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 20114033-8 2010 We also report a surprising finding that systemic application of MK-801 induced a similar age-related profile of changes in motor activity and c-Fos protein expression in the motor cortex but no c-Fos induction in the striatum. Dizocilpine Maleate 65-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-148 20331882-3 2010 The number of spinal c-Fos positive neurons of rats treated intrathecally with serotonin, noradrenaline or acetylcholine antagonists where evaluated to study the descending pathways activated by a surgical paw incision. Serotonin 79-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 20006681-8 2010 valine injection allowed earlier food intake compared with an i.p valine injection and increased the density of c-Fos-positive ependymal cells lining the third ventricle. Valine 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 20302612-3 2010 METHODS: First, rats subjected to either a supramaximal electrical stimulation or an injection of high-dose formalin in the hind limb were identified to have pain responses with behavioral evidence and spinal Fos-immunoreactive profiles. Formaldehyde 108-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 209-212 20059987-4 2010 We found that injections of METH (4x10 mg/kg, given at 2 h intervals) caused significant increases in c-fos and fra-2 expression which lasted from 30 min to 4 h. Pre-treatment with SCH23390, given 30 min before each METH injection, completely blocked METH-induced expression of c-fos, but only partially inhibited fra-2 mRNA expression. Methamphetamine 28-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 20059987-4 2010 We found that injections of METH (4x10 mg/kg, given at 2 h intervals) caused significant increases in c-fos and fra-2 expression which lasted from 30 min to 4 h. Pre-treatment with SCH23390, given 30 min before each METH injection, completely blocked METH-induced expression of c-fos, but only partially inhibited fra-2 mRNA expression. Methamphetamine 28-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 278-283 20059987-4 2010 We found that injections of METH (4x10 mg/kg, given at 2 h intervals) caused significant increases in c-fos and fra-2 expression which lasted from 30 min to 4 h. Pre-treatment with SCH23390, given 30 min before each METH injection, completely blocked METH-induced expression of c-fos, but only partially inhibited fra-2 mRNA expression. SCH 23390 181-189 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 20059987-4 2010 We found that injections of METH (4x10 mg/kg, given at 2 h intervals) caused significant increases in c-fos and fra-2 expression which lasted from 30 min to 4 h. Pre-treatment with SCH23390, given 30 min before each METH injection, completely blocked METH-induced expression of c-fos, but only partially inhibited fra-2 mRNA expression. SCH 23390 181-189 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 278-283 20059987-5 2010 These results were confirmed by Western blot analysis which showed METH-induced changes in c-Fos protein expression that were blocked by pretreatment with SCH23390. Methamphetamine 67-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 20059987-5 2010 These results were confirmed by Western blot analysis which showed METH-induced changes in c-Fos protein expression that were blocked by pretreatment with SCH23390. SCH 23390 155-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 19852984-9 2010 Robust increases in activity-related IEG expression in AID (c-fos and Arc) and Cg3 (c-fos) were observed following social interaction in saccharin-exposed rats, however, activity-related increases in IEG expression were not observed in fetal-ethanol-exposed rats in either region. Ethanol 242-249 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 20097261-1 2010 In the present study, we examined sex-specific changes in luteinizing hormone (LH) secretion and Fos expression in gonadotropin-releasing hormone (GnRH) neurons in response to naloxone in young (3 months old) and old (24 months old), gonadectomized male and female rats. Naloxone 176-184 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 20097261-2 2010 We revealed by immunocytochemistry that, regardless of age and sex, naloxone significantly increased the number of GnRH neurons expressing Fos, which was associated with increased LH secretion. Naloxone 68-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 20097261-2 2010 We revealed by immunocytochemistry that, regardless of age and sex, naloxone significantly increased the number of GnRH neurons expressing Fos, which was associated with increased LH secretion. Luteinizing Hormone 180-182 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-142 19056106-3 2010 At 60min after the EA treatment, the total number of c-fos-immunolabeled neurons in each region of the dorsal horn in lumbar segments (L4-5) was decreased by the BAPTA injection, particularly in the Nucleus proprius. 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid 162-167 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 19100597-0 2010 Single-dose and chronic corticosterone treatment alters c-Fos or FosB immunoreactivity in the rat cerebral cortex. Corticosterone 24-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 19100597-1 2010 The aim of this study was to examine the effects of single-dose and chronic corticosterone treatment on the inducible transcription factor c-Fos and FosB, and thereby to estimate the effects of high-doses of corticosterone on calcium-dependent neuronal responses in the rat cerebral cortex. Corticosterone 76-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 19100597-10 2010 It was found that a single-dose administration of corticosterone resulted in a significant, time-dependent increase of c-Fos protein immunoreactivity in the granule cell layer of the dentate gyrus, as well as in regions CA1 and CA3 of the hippocampus 12 and 24h post-injection with respect to control animals. Corticosterone 50-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 19100597-18 2010 The present data suggest that single-dose corticosterone treatment increases immunoreactivity of c-Fos protein in a time-dependent manner, 12 and 24h post-injection in the rat hippocampus and the neocortex, whereas chronic corticosterone treatment influences FosB immunoreactivity, primarily in the dentate gyrus. Corticosterone 42-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 19515121-8 2010 Anandamide also proved effective in preventing nitroglycerin-induced activation (c-Fos) of neurons in the nucleus trigeminalis caudalis. anandamide 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 19515121-8 2010 Anandamide also proved effective in preventing nitroglycerin-induced activation (c-Fos) of neurons in the nucleus trigeminalis caudalis. Nitroglycerin 47-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 19525133-4 2010 In this study, a noxious injection of formalin was given into the lower lip of female rats, thereby activating nociceptive neurons in the trigeminal subnucleus caudalis as demonstrated by immunohistochemical labeling of Fos. Formaldehyde 38-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 220-223 19833107-0 2010 Visualizing acute pain-morphine interaction in descending monoamine nuclei with Fos. Morphine 23-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 19931852-6 2010 RESULTS: Fluoxetine robustly potentiated methylphenidate-induced expression of the transcription factors c-fos and zif 268 throughout the striatum and to some degree in the nucleus accumbens. Fluoxetine 9-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 19931852-6 2010 RESULTS: Fluoxetine robustly potentiated methylphenidate-induced expression of the transcription factors c-fos and zif 268 throughout the striatum and to some degree in the nucleus accumbens. Methylphenidate 41-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 20028457-7 2010 Nicotine self-administration also decreased footshock-induced c-Fos expression in the nucleus of the solitary tract-A2/C2 catecholaminergic neurons that project to the PVN. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 19915516-0 2010 Effect of complete maternal and littermate deprivation on morphine-induced Fos-immunoreactivity in the adult male rat brain. Morphine 58-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 19915516-7 2010 Specifically, relative to MR rats, AR rats showed significantly greater morphine-induced Fos-immunoreactivity in brain regions associated with the mesocorticolimbic "reward" pathway. Morphine 72-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 19774673-6 2010 Most apparent activation of single Fos, Fos/OXY, and Fos/AVP cells was induced by clozapine and olanzapine; effects of risperidone and haloperidol were substantially lower; no colocalizations were revealed in naive or vehicle treated control rats. Clozapine 82-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 19774673-6 2010 Most apparent activation of single Fos, Fos/OXY, and Fos/AVP cells was induced by clozapine and olanzapine; effects of risperidone and haloperidol were substantially lower; no colocalizations were revealed in naive or vehicle treated control rats. Clozapine 82-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 19774673-6 2010 Most apparent activation of single Fos, Fos/OXY, and Fos/AVP cells was induced by clozapine and olanzapine; effects of risperidone and haloperidol were substantially lower; no colocalizations were revealed in naive or vehicle treated control rats. Olanzapine 96-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 19774673-6 2010 Most apparent activation of single Fos, Fos/OXY, and Fos/AVP cells was induced by clozapine and olanzapine; effects of risperidone and haloperidol were substantially lower; no colocalizations were revealed in naive or vehicle treated control rats. Olanzapine 96-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 19774673-6 2010 Most apparent activation of single Fos, Fos/OXY, and Fos/AVP cells was induced by clozapine and olanzapine; effects of risperidone and haloperidol were substantially lower; no colocalizations were revealed in naive or vehicle treated control rats. Olanzapine 96-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-43 19800921-5 2010 RESULTS: Confirming previous results systemic administration of CBD (10mg/kg) decreased contextual fear and associated c-Fos expression in the prefrontal cortex (prelimbic and infralimbic regions). Cannabidiol 64-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 20039957-10 2010 RESULTS: Chronic paracetamol exposure led to an increase in CSD frequency and CSD-evoked Fos expression in cerebral cortex indicating the increase in neuronal excitability. Acetaminophen 17-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 19531280-5 2010 The DCE was selectively associated with an increase in Fos immunoreactivity in the medial prefrontal cortex (mPFC), an increase that was not observed in the presence of atropine. ethylene dichloride 4-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 19531280-6 2010 Here, we set out to test the actions of typical, atypical and potential antipsychotics on atropine-induced disruption of the DCE and the related mPFC Fos-immunoreactivity profile. Atropine 90-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 150-153 19531280-9 2010 Compared to contingent control groups, an increased level of Fos immunoreactivity within the mPFC was observed only with doses that reversed atropine-induced disruption of the DCE. Atropine 141-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 19531280-9 2010 Compared to contingent control groups, an increased level of Fos immunoreactivity within the mPFC was observed only with doses that reversed atropine-induced disruption of the DCE. ethylene dichloride 176-179 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 19891980-9 2010 The number of carrageenan-induced Fos-like immunoreactive profiles was less in rosiglitazone-treated rats as compared to vehicle controls. Carrageenan 14-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 19891980-9 2010 The number of carrageenan-induced Fos-like immunoreactive profiles was less in rosiglitazone-treated rats as compared to vehicle controls. Rosiglitazone 79-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 19924112-8 2010 The KOR antagonist nor-binaltorphimine produced differential effects on the number of c-fos-positive profiles in the piriform cortex and nucleus accumbens shell between SD and WKY rats. norbinaltorphimine 19-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 20026253-3 2010 Immunohistochemical double-labeling technique with Fos was used to examine if an intraplantar formalin injection, an acute noxious input, changed the effect of morphine on dopaminergic neurons of the ventral tegmental area (VTA), and serotonergic neurons of the dorsal raphe nucleus (DR). Formaldehyde 94-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 20026253-5 2010 Following morphine injection, there was an increase in the number of dopaminergic neurons in the VTA with Fos immunolabeling. Morphine 10-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 20026253-7 2010 In contrast, the number of serotonergic neurons containing Fos was increased in the morphine/formalin group compared to all other groups and this effect was topographically selective for the dorsal area of the DR at mid rostro-caudal levels. Morphine 84-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 20026253-7 2010 In contrast, the number of serotonergic neurons containing Fos was increased in the morphine/formalin group compared to all other groups and this effect was topographically selective for the dorsal area of the DR at mid rostro-caudal levels. Formaldehyde 93-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-62 19815001-3 2010 Fos expression in LH orexin neurons varied in proportion to conditioned place preference (CPP) for morphine, cocaine, or food. Morphine 99-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 19815001-3 2010 Fos expression in LH orexin neurons varied in proportion to conditioned place preference (CPP) for morphine, cocaine, or food. Cocaine 109-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 19774673-6 2010 Most apparent activation of single Fos, Fos/OXY, and Fos/AVP cells was induced by clozapine and olanzapine; effects of risperidone and haloperidol were substantially lower; no colocalizations were revealed in naive or vehicle treated control rats. Clozapine 82-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 19520080-6 2010 CLS induced Fos in hypothalamic and limbic structures, including the nucleus accumbens, piriform cortex, arcuate nucleus, and dorsomedial portion of the ventromedial hypothalamus, compared to no stimulation. calusterone 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-15 19520080-7 2010 However, distributed CLS induced more Fos in the medial preoptic area than continuous CLS or no stimulation. calusterone 21-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 19520080-8 2010 In contrast, continuous CLS induced more Fos in the posteroventral medial amygdala compared to no stimulation. calusterone 24-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 19520080-9 2010 These data indicate that CLS induces a reward state in the rat and a pattern of Fos activation in regions of the brain that process genitosensory input, incentive salience, and reward. calusterone 25-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 19943848-9 2010 Our data strongly indicate that Galpha(q/11)-mediated ERK1/2 activation is essential for expression of Fos upon illumination of melanopsin-expressing PC12 cells. galpha 32-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-106 19747496-3 2010 We herein investigated whether the blockade of TTX-sensitive channels could reduce pain-related behaviors and evoked c-Fos immunoreactivity in rats with neuropathic pain produced by chronic unilateral constriction injury to either the sciatic nerve or the infraorbital nerve. Tetrodotoxin 47-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 19747496-6 2010 In sciatic nerve-ligated rats, TTX administration prevented the increased c-Fos immunoreactivity occurring in the dorsal horn of the lumbar cord and some supraspinal areas in response to light mechanical stimulation of the nerve-injured hindpaw. Tetrodotoxin 31-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-79 20101163-15 2010 Expression of the c-Fos protein was significantly increased in the medial vestibular nuclei and inferior vestibular nuclei at 1, 2, and 6 hours after UL, and the expression was significantly decreased in animals that were pretreated with MK-801 (p < 0.01). Dizocilpine Maleate 238-244 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 20004702-4 2010 The intraperitoneal administration of 1mg/kg of SB 243213 or SER-082, but not mianserin, enhanced Fos-immunoreactive cells in the subthalamic nucleus and the striatum, primarily its medial portion. Antimony 48-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 20004702-4 2010 The intraperitoneal administration of 1mg/kg of SB 243213 or SER-082, but not mianserin, enhanced Fos-immunoreactive cells in the subthalamic nucleus and the striatum, primarily its medial portion. Serine 61-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 19766144-6 2010 To elucidate neural correlates of our behavioral results, we have used c-fos expression to provide a global map of neuronal activity, with single cell resolution, in the olfactory bulb of D- and L-carvone exposed animals. d- and l-carvone 188-204 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 19783484-4 2010 In response to intermittent intravenous injections of KCN, a significant increase in the number of Fos-ir neurons was observed specifically in the lateral intermediate commissural NTS [(LI)NTS (82+/-9 vs. 174+/-16, cell number mean per section)] and lateral caudal commissural NTS [(LC)NTS (71+/-9 vs. 199+/-18, cell number mean per section)]. Potassium Cyanide 54-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 19944746-0 2010 The effects of glycine transporter I inhibitor, N-methylglycine (sarcosine), on ketamine-induced alterations in sensorimotor gating and regional brain c-Fos expression in rats. Sarcosine 65-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-156 19944746-0 2010 The effects of glycine transporter I inhibitor, N-methylglycine (sarcosine), on ketamine-induced alterations in sensorimotor gating and regional brain c-Fos expression in rats. Ketamine 80-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-156 19944746-3 2010 We assessed the antipsychotic potential of sarcosine by comparing the abilities of sarcosine and clozapine to restore the prepulse inhibition (PPI) deficit, hyperlocomotion and regional brain c-Fos expression changes caused by an NMDAR antagonist, ketamine. Clozapine 97-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 192-197 19944746-7 2010 However, in the olfactory bulb, sarcosine, but not clozapine, significantly reduced the ketamine-induced increase in c-Fos expression. Sarcosine 32-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 19944746-7 2010 However, in the olfactory bulb, sarcosine, but not clozapine, significantly reduced the ketamine-induced increase in c-Fos expression. Ketamine 88-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 19857504-12 2010 We found intense NADPHd activity in PMV neurons, some of which coexpressed Fos-ir after exposure to both odors. nadphd 17-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 19782661-7 2010 SB 242084 also completely blocked the LPS-induced increases in c-Fos expression in the paraventricular nucleus, central nucleus of the amygdala, nucleus tractus solitarii, median raphe nucleus and dorsal raphe nucleus and partially blocked it in the A1 noradrenergic area of the ventrolateral medulla and the raphe pallidus nucleus. 6-chloro-5-methyl-1-((2-(2-methylpyrid-3-yloxy)pyrid-5-yl)carbamoyl)indoline 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 19833107-5 2010 However, the percentage of noradrenergic neurons in the A7 and A5 cell groups that contained Fos was significantly increased in the morphine/formalin group compared to all other groups, while no differences were found in serotonin cells in the NRM. Morphine 132-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 19833107-5 2010 However, the percentage of noradrenergic neurons in the A7 and A5 cell groups that contained Fos was significantly increased in the morphine/formalin group compared to all other groups, while no differences were found in serotonin cells in the NRM. Formaldehyde 141-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-96 19833107-0 2010 Visualizing acute pain-morphine interaction in descending monoamine nuclei with Fos. monoamine 58-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 19923355-4 2010 After injection of vehicle into the dorsomedial hypothalamus, injection of muscimol into the medial preoptic area elicited marked increases in heart rate, arterial pressure, body temperature, plasma ACTH, and locomotor activity and also increased c-Fos expression in the hypothalamic paraventricular nucleus, a region known to control the release of ACTH from the adenohypophysis. Muscimol 75-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 247-252 19852649-2 2010 Also, the effect of morphine on Fos immunoreactivity (Fos-IR) was investigated in these nuclei. Morphine 20-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-35 19852649-2 2010 Also, the effect of morphine on Fos immunoreactivity (Fos-IR) was investigated in these nuclei. Morphine 20-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 19852649-10 2010 Furthermore, pretreatment with morphine in the 70g group reduced Fos-IR in these regions. Morphine 31-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 20074225-1 2010 We report that satiation evokes neuronal activity in the ventral subdivision of the hypothalamic dorsomedial nucleus (DMH) as indicated by increased c-fos expression in response to refeeding in fasted rats. Dimenhydrinate 118-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 149-154 19889861-12 2010 OCT injection increased single c-Fos-labeled cell counts in the MnPN, but not in the VLPO. maxacalcitol 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 20074225-7 2010 In accord with these changes, the number of Fos-expressing neurons following refeeding declined in the ipsilateral but remained high in the contralateral DMH. Dimenhydrinate 154-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 20074225-8 2010 However, the Fos response in the ventral DMH was not attenuated following chemical lesion (neonatal monosodium glutamate treatment) of the hypothalamic arcuate nucleus, another possible source of DMH inputs. Dimenhydrinate 41-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 20213575-9 2010 Phenylephrine activated the c-fos gene expression. Phenylephrine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-33 21324267-3 2010 Diisopropylfluorophosphatetreated animals exhibited frequent yawning, significant inhibition of acetylcholinesterase staining and upregulation of c-fos mRNA, but not the epileptic seizures or significant change of Syt4 mRNA levels. diisopropylfluorophosphatetreated 0-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 146-151 19794406-0 2010 Fos after single and repeated self-administration of cocaine and saline in the rat: emphasis on the Basal forebrain and recalibration of expression. Cocaine 53-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 19794406-0 2010 Fos after single and repeated self-administration of cocaine and saline in the rat: emphasis on the Basal forebrain and recalibration of expression. Sodium Chloride 65-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 19794406-2 2010 We evaluated a large sample of brain structures, particularly ones comprising basal forebrain macrosystems, and determined in which the immediate-early gene product, Fos, is expressed following a single and repeated self-administrations of cocaine. Cocaine 240-247 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-169 19794406-4 2010 Fos expression was mainly enhanced by acutely self-administered cocaine in basal ganglia output and intrinsic structures and the intermediate nucleus of lateral septum, medial division of the central amygdaloid nucleus and zona incerta, but, in contrast, was sensitized in these structures after repeated administrations. Cocaine 64-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 19794406-6 2010 Thus, self-administered cocaine mainly elicits Fos expression, which persists or increases with repeated administrations in some structures, but declines in others. Cocaine 24-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 19794406-8 2010 Similar spatiotemporal patterns of cocaine- or saline-elicited Fos expression characterize functionally related clusters of structures, such as, eg, basal ganglia input structures, basal ganglia output structures, extended amygdala and structures in the brainstem to which forebrain macrosystems project. Cocaine 35-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 19794406-8 2010 Similar spatiotemporal patterns of cocaine- or saline-elicited Fos expression characterize functionally related clusters of structures, such as, eg, basal ganglia input structures, basal ganglia output structures, extended amygdala and structures in the brainstem to which forebrain macrosystems project. Sodium Chloride 47-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 19800912-0 2010 D1 priming enhances both D1- and D2-mediated rotational behavior and striatal Fos expression in 6-hydroxydopamine lesioned rats. Oxidopamine 96-113 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 19800912-5 2010 6-OHDA rats challenged with SKF38393 (1mg/kg) showed no contralateral rotational behavior, but robust striatal Fos expression in D1-primed animals. Oxidopamine 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 19800912-5 2010 6-OHDA rats challenged with SKF38393 (1mg/kg) showed no contralateral rotational behavior, but robust striatal Fos expression in D1-primed animals. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 28-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-114 19800912-6 2010 Challenge with SKF38393 (10mg/kg) led to pronounced contralateral rotational behavior and striatal Fos expression in all priming groups - with the largest behavioral response in D1- and D2-primed rats. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 15-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 19800912-7 2010 Quinpirole challenge (0.25mg/kg) led to robust contralateral rotational behavior and striatal Fos expression in D1-primed animals, but only mild rotational behavior and baseline levels of striatal Fos expression in D2-primed animals. Quinpirole 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 19800912-7 2010 Quinpirole challenge (0.25mg/kg) led to robust contralateral rotational behavior and striatal Fos expression in D1-primed animals, but only mild rotational behavior and baseline levels of striatal Fos expression in D2-primed animals. Quinpirole 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 197-200 19800912-8 2010 These data suggest that D1- or D2-priming enhances rotational behavior following challenge with D1 or D2 agonist, but only D1-priming enhances D1- and D2-mediated striatal Fos expression in 6-OHDA rats. Oxidopamine 190-196 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 172-175 20213575-10 2010 Hyperglycemia augmented the phenylephrine-induced c-fos gene expression synergistically in a dose dependent manner. Phenylephrine 28-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 19782054-9 2009 injection of formalin (1%) produced significantly fewer nocifensive behaviors and expression of c-Fos protein in the spinal dorsal horn, confirming the hypoalgesic status in the inflamed site. Formaldehyde 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 20034457-3 2010 The acupuncture treatment was performed for 1 minute once a day for 3 days of withdrawal period and its effect on morphine-induced changes of locomotor activity and Fos expression was examined. Morphine 114-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-168 20034457-4 2010 RESULTS: The acupuncture stimulation to HT7 significantly suppressed the morphine-induced increases in the locomotor activity and Fos expression in the nucleus accumbens and striatum, as compared to the controls of non-acupoint or the acupoint on other meridian. Morphine 73-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-133 19772897-4 2009 In intact males, MK801 triggered a c-Fos induction that remained high for more than 24 h in selected layers of the retrosplenial, somatosensory and entorhinal cortices. Dizocilpine Maleate 17-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 19733634-3 2009 Results showed that the Fos immunoreactivity (Fos-ir) expression in forebrain areas such as subfornical organ (SFO), paraventricular hypothalamic nuclei (PVN), supraoptic nucleus (SON) and organum vasculosum laminae terminalis (OVLT) all increased significantly and that the levels of ANG I, ANG II and ALD also increased in plasma and forebrain in rats fed with low sodium diet. Sodium 367-373 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 19733634-3 2009 Results showed that the Fos immunoreactivity (Fos-ir) expression in forebrain areas such as subfornical organ (SFO), paraventricular hypothalamic nuclei (PVN), supraoptic nucleus (SON) and organum vasculosum laminae terminalis (OVLT) all increased significantly and that the levels of ANG I, ANG II and ALD also increased in plasma and forebrain in rats fed with low sodium diet. Sodium 367-373 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 19864960-0 2009 Caffeine elicits c-Fos expression in horizontal diagonal band cholinergic neurons. Caffeine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 19864960-5 2009 Caffeine significantly increased c-Fos expression in cholinergic neurons of the horizontal limb of the diagonal band of Broca but not other basal forebrain regions such as the medial septum or substantia innominata. Caffeine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 19850746-3 2009 In c-fos-mRFP1 transgenic rats, 90 min after hypertonic saline ip administration, nuclear mRFP1 fluorescence was observed abundantly in brain regions known to be osmosensitive, namely the median preoptic nucleus, organum vasculosum lamina terminalis, supraoptic nucleus, paraventricular nucleus, and subfornical organ. Sodium Chloride 56-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 3-8 19822518-11 2009 As reported previously for electrical stimulation, 500 mum ATP significantly increased mRNA expression for both c-fos and interleukin 6. Adenosine Triphosphate 59-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-117 19782659-0 2009 Damage to the central amygdala produces differential encephalic c-fos expression in the water deprivation-partial rehydration protocol. Water 88-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 19699782-0 2009 The anterior cingulate cortex is a target structure for the anxiolytic-like effects of benzodiazepines assessed by repeated exposure to the elevated plus maze and Fos immunoreactivity. Benzodiazepines 87-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 163-166 19699782-9 2009 Fos immunohistochemistry showed that midazolam injections were associated with a distinct pattern of action when administered before the test or retest session, and the anterior cingulate cortex, area 1 (Cg1), was the only structure targeted by the benzodiazepine in both situations. Midazolam 37-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 19699782-9 2009 Fos immunohistochemistry showed that midazolam injections were associated with a distinct pattern of action when administered before the test or retest session, and the anterior cingulate cortex, area 1 (Cg1), was the only structure targeted by the benzodiazepine in both situations. Benzodiazepines 249-263 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 19782067-7 2009 Moreover, minocycline partially inhibited BmK venom-induced spinal c-Fos expression but lack of effects on BmK venom-induced paw edema. Minocycline 10-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 19710391-6 2009 We investigated the role of the lateral parabrachial nucleus (lPBN), a major visceral afferent link between the hindbrain and forebrain, in cisplatin-induced c-Fos expression and pica. Cisplatin 140-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 19710391-7 2009 Injection of cisplatin (6 mg/kg ip) produced c-Fos expression in the ventrolateral (external) lPBN, a region receiving viscerosensory input. Cisplatin 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 19737844-6 2009 In comparison with vehicle infusion or the non-injected side of the face, capsaicin significantly increased the expression of the activation markers Fos in the spinal trigeminal nucleus and phosphorylated extracellular signal-regulated kinase in the trigeminal ganglion. Capsaicin 74-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 149-152 19737844-7 2009 Pre-treatment with olcegepant (900 microg/kg) inhibited the capsaicin-induced expression of Fos throughout the spinal trigeminal nucleus by 57%. olcegepant 19-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 19737844-7 2009 Pre-treatment with olcegepant (900 microg/kg) inhibited the capsaicin-induced expression of Fos throughout the spinal trigeminal nucleus by 57%. Capsaicin 60-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 19845681-4 2009 The activity of DRN GABA neurones was assessed using double-label immunohistochemical measurements of Fos and glutamate decarboxylase (GAD). gamma-Aminobutyric Acid 20-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 19778539-5 2009 Fos/OT neurons in the posterior parvocellular subdivision of the PVN were increased after refeeding, with a higher number in the ADX group, compared with sham and ADX+corticosterone (B) groups, with no difference in the medial parvocellular and magnocellular subdivisions of the PVN. Corticosterone 167-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 18754816-2 2009 The present study was undertaken to test the hypothesis that PPARalpha activator fenofibrate plays a key role in left ventricular hypertrophic remodelling via the formation of c-fos/c-jun heterodimers in spontaneous hypertensive rats (SHRs). Fenofibrate 81-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 176-181 18754816-6 2009 Similarly, the excessive collagen deposition and the up-regulation of collagen I, collagen III, c-fos and c-jun seen in SHRs receiving saline were significantly attenuated in SHRs receiving fenofibrate. Sodium Chloride 135-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 18754816-6 2009 Similarly, the excessive collagen deposition and the up-regulation of collagen I, collagen III, c-fos and c-jun seen in SHRs receiving saline were significantly attenuated in SHRs receiving fenofibrate. Fenofibrate 190-201 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 96-101 18754816-7 2009 In addition, fenofibrate markedly decreased the expression of AP-1 and c-fos/c-jun heterodimers (P < 0.01). Fenofibrate 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 18754816-8 2009 These results demonstrated that PPARalpha activator fenofibrate may exert a protective effect on cardiac remodelling in SHRs by decreasing the expression of c-fos and c-jun and suppressing the formation of c-fos/c-jun heterodimers, which may further inhibit transcription of the downstream genes involved in the pathogenesis of left ventricular hypertrophy induced by hypertension. Fenofibrate 52-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-162 18754816-8 2009 These results demonstrated that PPARalpha activator fenofibrate may exert a protective effect on cardiac remodelling in SHRs by decreasing the expression of c-fos and c-jun and suppressing the formation of c-fos/c-jun heterodimers, which may further inhibit transcription of the downstream genes involved in the pathogenesis of left ventricular hypertrophy induced by hypertension. Fenofibrate 52-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 206-211 19699734-9 2009 These results suggested that curcumin suppressed the increased bone resorptive activity through the prevention of osteoclastogenesis associated with inhibition of the expression of c-fos and c-jun in the diabetic rats. Curcumin 29-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 181-186 19695778-7 2009 Selectively blocking either NR2A or NR2B subunit in the rACC abolished the acquisition of F-CPA and formalin nociceptive conditioning-induced Fos expression, but it did not affect formalin acute nociceptive behaviors and non-nociceptive fear stimulus-induced CPA. Formaldehyde 100-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-145 19711055-0 2009 Nicotine-conditioned place preference induced CREB phosphorylation and Fos expression in the adult rat brain. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 19711055-6 2009 During nicotine preference and reinstatement behaviors, a significant increase of both pCREB and Fos protein expression occurs in the nucleus accumbens (NAc) and ventral tegmental area (VTA) and also in the prefrontal cortex (PFC), dorsal striatum (DStr), amygdala, and hippocampus. Nicotine 7-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-100 19711055-9 2009 CONCLUSION: The results indicate that the phosphorylation of CREB and expression of Fos protein, as indicators of neural activity, accompany the acquisition and maintenance of nicotine-induced CPP but not CPA in mesolimbic areas (NAc, VTA, PFC, and DStr) as well as in memory consolidation structures (hippocampus and amygdala) and nicotinic receptor are involved in this process. Nicotine 176-184 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 19695244-3 2009 F15063 (10 mg/kg) and clozapine (10 mg/kg), but not haloperidol (0.63 mg/kg), induced c-fos and fosB mRNA expression in prefrontal cortex, a region associated with control of cognition and negative symptoms of schizophrenia. N-((2,2-dimethyl-2,3-dihydrobenzofuran-7-yloxy)ethyl)-3-(cyclopent-1-enyl)benzylamine 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 19683523-7 2009 Moreover, c-Fos expression was induced in the PVN after naloxone-precipitated morphine withdrawal. Naloxone 56-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-15 19683523-7 2009 Moreover, c-Fos expression was induced in the PVN after naloxone-precipitated morphine withdrawal. Morphine 78-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-15 19695244-3 2009 F15063 (10 mg/kg) and clozapine (10 mg/kg), but not haloperidol (0.63 mg/kg), induced c-fos and fosB mRNA expression in prefrontal cortex, a region associated with control of cognition and negative symptoms of schizophrenia. Clozapine 22-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 19695244-4 2009 In striatum, only c-fos, fosB, junB and nur77 were induced by clozapine whereas all IEG mRNAs were increased by haloperidol and F15063 (from 2.5 mg/kg) with similar high efficacy despite a total absence of F15063-induced catalepsy. Clozapine 62-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 19738498-5 2009 The intragastric administration of 3 mg/kg of donepezil significantly increased the plasma adrenocorticotropic hormone levels and c-Fos expression in the paraventricular nucleus, and decreased the food intake on the first day. Donepezil 46-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 19535786-9 2009 FOS induction in hypothalamic gonadotropic (GnRH) neurons after hormone priming was impaired in the EB- and PPT-treated groups but neither of the BPA-treated groups. 4,4',4''-(4-propyl-((1)H)-pyrazole-1,3,5-triyl) tris-phenol 108-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 19535786-9 2009 FOS induction in hypothalamic gonadotropic (GnRH) neurons after hormone priming was impaired in the EB- and PPT-treated groups but neither of the BPA-treated groups. bisphenol A 146-149 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 19735534-7 2009 The number of c-fos+ neurons in superficial lamina of TNC was significantly higher in the Saharan dust group (32.9 +/- 5.3, P = 0.0001) compared with dust-free air (11.02 +/- 2.7) or Co(60)-treated Saharan dust groups (15.01 +/- 2.4). co(60) 183-189 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 19916832-11 2009 However, the number of cells expressing cFos in the vSPVZ was positively correlated with general activity during the subjective day relative to the subjective night when the animals were switched to DD, and this pattern persisted when a LD cycle was reinstated. Fumigant 93 199-201 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-44 19713157-0 2009 Mapping of c-Fos expression in the rat brain during the evolution of pentylenetetrazol-kindled seizures. Pentylenetetrazole 69-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 19713157-1 2009 c-Fos protein immunocytochemistry was used to map the brain structures recruited during the evolution of seizures that follows repeated administration of a subconvulsive dose (35mg/kg, ip) of pentylenetetrazol in rats. Pentylenetetrazole 192-209 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 19665463-0 2009 Vaginocervical stimulation induces Fos in glutamate neurons in the ventromedial hypothalamus: attenuation by estrogen and progesterone. Glutamic Acid 42-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 19665463-0 2009 Vaginocervical stimulation induces Fos in glutamate neurons in the ventromedial hypothalamus: attenuation by estrogen and progesterone. Progesterone 122-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 19665463-5 2009 The present experiment examined the ability of VCS to induce Fos in glutamate neurons in the VMH of ovariectomized rats under 4 hormonal regimens: oil, EB alone, P alone, or EB+P, following 1 or 50 distributed VCSs administered with a lubricated glass rod over the course of 1 h. Treatment with EB or P alone significantly reduced the number of glutamate neurons activated by 1 VCS, with P being more effective than EB. Glutamic Acid 68-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 19563846-1 2009 In the present study, on rats, a quantitative analysis of Fos protein immunohistochemistry was performed as a way of investigating the effects of inhalation of green odor (a mixture of equal amounts of trans-2-hexenal and cis-3-hexenol) on the neuronal activations in stress-related forebrain regions induced by acute and repeated stress. 3-hexen-1-ol 222-235 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 19467297-6 2009 Repeated administration of 1.0 mg/kg pimozide induced tremulous jaw movements and increased ventrolateral striatal c-Fos expression, while administration of 20.0 mg/kg of the atypical antipsychotic quetiapine did not. Pimozide 37-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-120 19467297-8 2009 Pimozide-induced increases in ventrolateral striatal c-Fos expression were reduced by a behaviorally effective dose of KW 6002, but c-Fos expression in pimozide-treated rats was actually increased by tropicamide. Pimozide 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 19467297-8 2009 Pimozide-induced increases in ventrolateral striatal c-Fos expression were reduced by a behaviorally effective dose of KW 6002, but c-Fos expression in pimozide-treated rats was actually increased by tropicamide. Pimozide 152-160 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 19467297-8 2009 Pimozide-induced increases in ventrolateral striatal c-Fos expression were reduced by a behaviorally effective dose of KW 6002, but c-Fos expression in pimozide-treated rats was actually increased by tropicamide. Tropicamide 200-211 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 19467297-9 2009 These results indicate that two different drug manipulations that act to reduce tremulous jaw movements can have different effects on DA antagonist-induced c-Fos expression, suggesting that adenosine A(2A) antagonism and muscarinic receptor antagonism exert their motor effects by acting on different striatal circuits. amsonic acid 134-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 19467297-9 2009 These results indicate that two different drug manipulations that act to reduce tremulous jaw movements can have different effects on DA antagonist-induced c-Fos expression, suggesting that adenosine A(2A) antagonism and muscarinic receptor antagonism exert their motor effects by acting on different striatal circuits. adenosine a 190-201 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-161 19291390-10 2009 ATIP or IDX only partially reduced Fos in SAL or XYL treated di/di rats. atipamezole 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 19463813-6 2009 We found that the ionotropic glutamate agonists (NMDA plus AMPA) caused an approximately 6-fold increase of c-fos gene expression in the SON, and some, but not all, G-coupled protein receptor agonists (e.g., phenylephrine, senktide, a NK-3-receptor agonist, and alpha-MSH) increased the c-fos gene expression in the SON from between 1.5 to 2-fold of the control SONs. Glutamic Acid 29-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 19463813-6 2009 We found that the ionotropic glutamate agonists (NMDA plus AMPA) caused an approximately 6-fold increase of c-fos gene expression in the SON, and some, but not all, G-coupled protein receptor agonists (e.g., phenylephrine, senktide, a NK-3-receptor agonist, and alpha-MSH) increased the c-fos gene expression in the SON from between 1.5 to 2-fold of the control SONs. Glutamic Acid 29-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 287-292 19463813-6 2009 We found that the ionotropic glutamate agonists (NMDA plus AMPA) caused an approximately 6-fold increase of c-fos gene expression in the SON, and some, but not all, G-coupled protein receptor agonists (e.g., phenylephrine, senktide, a NK-3-receptor agonist, and alpha-MSH) increased the c-fos gene expression in the SON from between 1.5 to 2-fold of the control SONs. N-Methylaspartate 49-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 19463813-6 2009 We found that the ionotropic glutamate agonists (NMDA plus AMPA) caused an approximately 6-fold increase of c-fos gene expression in the SON, and some, but not all, G-coupled protein receptor agonists (e.g., phenylephrine, senktide, a NK-3-receptor agonist, and alpha-MSH) increased the c-fos gene expression in the SON from between 1.5 to 2-fold of the control SONs. N-Methylaspartate 49-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 287-292 19463813-6 2009 We found that the ionotropic glutamate agonists (NMDA plus AMPA) caused an approximately 6-fold increase of c-fos gene expression in the SON, and some, but not all, G-coupled protein receptor agonists (e.g., phenylephrine, senktide, a NK-3-receptor agonist, and alpha-MSH) increased the c-fos gene expression in the SON from between 1.5 to 2-fold of the control SONs. alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid 59-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 19463813-6 2009 We found that the ionotropic glutamate agonists (NMDA plus AMPA) caused an approximately 6-fold increase of c-fos gene expression in the SON, and some, but not all, G-coupled protein receptor agonists (e.g., phenylephrine, senktide, a NK-3-receptor agonist, and alpha-MSH) increased the c-fos gene expression in the SON from between 1.5 to 2-fold of the control SONs. alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid 59-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 287-292 19463813-6 2009 We found that the ionotropic glutamate agonists (NMDA plus AMPA) caused an approximately 6-fold increase of c-fos gene expression in the SON, and some, but not all, G-coupled protein receptor agonists (e.g., phenylephrine, senktide, a NK-3-receptor agonist, and alpha-MSH) increased the c-fos gene expression in the SON from between 1.5 to 2-fold of the control SONs. Phenylephrine 208-221 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 19484338-0 2009 c-Fos expression in the supraoptic nucleus is the most intense during different durations of restraint water-immersion stress in the rat. Water 103-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 19065454-6 2009 CORT containing saline partially normalized basal and restraint-induced ACTH secretion and restraint-induced AVP hnRNA, c-fos mRNA, and zif268 mRNA in the PVN in ADX rats. Sodium Chloride 16-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 19463915-4 2009 of two NSAIDs with comparable spinal pharmacokinetics, ketoprofen (moderately preferential for COX-1) and parecoxib (selective COX-2), on the activation of spinal nociceptive neurons (measured as c-Fos expression) induced by carrageenan-induced acute inflammation in the rat paw. parecoxib 106-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 198-201 19463915-6 2009 Post-treatment with ketoprofen produced inhibition of c-Fos but parecoxib did not have any significant effect. Ketoprofen 20-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 19463915-8 2009 Paradoxically, pre-treatment with ketoprofen produced a greater inhibition of c-Fos expression than with parecoxib, in all lamina of ipsilateral dorsal horn of the lumbar spinal cord. Ketoprofen 34-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 19708041-0 2009 Regional Fos expression induced by morphine withdrawal in the 7-day-old rat. Morphine 35-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 19533625-0 2009 c-Fos expression associated with reinstatement of cocaine-seeking behavior by response-contingent conditioned cues. Cocaine 50-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 19533625-10 2009 We also observed a correlation between cocaine-seeking behavior and Fos in the agranular insula (AgI) and basolateral amygdala (BLA). Cocaine 39-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 19477242-0 2009 Nutritional status modulates behavioural and olfactory bulb Fos responses to isoamyl acetate or food odour in rats: roles of orexins and leptin. isoamyl acetate 77-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 19595675-8 2009 Fos-immunoreactive neurons were observed to be closely apposed by BDA-labelled fibres and terminal boutons. biotinylated dextran amine 66-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 19686468-8 2009 Furthermore, in a separate experiment, a prior experience with controllable stress led to potentiation of Fos expression in retrogradely labeled PL neurons in response to an uncontrollable stressor 1 week later. pl 145-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 19559985-0 2009 Pre-treatment with lidocaine suppresses ectopic discharges and attenuates neuropeptide Y and c-Fos expressions in the rat cuneate nucleus following median nerve transection. Lidocaine 19-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 19559985-8 2009 Lidocaine pre-treatment also attenuated the number of injury-induced NPY-LI fibers and c-Fos-LI neurons within the CN in a dose-dependent manner. Lidocaine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 19481580-0 2009 Methadone is substantially less effective than morphine in modifying locomotor and brain Fos responses to subsequent methadone challenge in rats. Methadone 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 19481580-0 2009 Methadone is substantially less effective than morphine in modifying locomotor and brain Fos responses to subsequent methadone challenge in rats. Morphine 47-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 19481580-0 2009 Methadone is substantially less effective than morphine in modifying locomotor and brain Fos responses to subsequent methadone challenge in rats. Methadone 117-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 19481580-6 2009 Sensitization of Fos response was found in a few regions of the morphine-treated rats. Morphine 64-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 19481580-8 2009 The rats given the lower methadone dose treatment showed a weak while widespread tendency for an opposite change, which reached significance in cingulate cortex layer II/III and resulted in significant differences in Fos response between these rats and the morphine-treated rats in most regions studied. Methadone 25-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 217-220 19481580-8 2009 The rats given the lower methadone dose treatment showed a weak while widespread tendency for an opposite change, which reached significance in cingulate cortex layer II/III and resulted in significant differences in Fos response between these rats and the morphine-treated rats in most regions studied. Morphine 257-265 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 217-220 19422884-8 2009 Pretreatment with CTAP prevented the increase in c-Fos translation in each of these areas. CTAP 18-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 19597526-8 2009 RESULTS: Cardiomyocytes cultured in a high concentration of glucose showed an increased pulsatile frequency and cellular volume, as well as a higher expression of PKC, NF-kappaB, TNF-alpha, and c-fos compared with the control group. Glucose 60-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 194-199 19597526-10 2009 Diabetic rats showed relative cardiac hypertrophy and a higher expression of PKC, NF-kappaB, TNF-alpha, and c-fos; treatment with breviscapine could ameliorate these changes in diabetic cardiomyopathy. breviscapine 130-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 19597526-11 2009 CONCLUSION: Breviscapine prevented cardiac hypertrophy in diabetic rats by inhibiting the expression of PKC, which may have a protective effect in the pathogenesis of diabetic cardiomyopathy via the PKC/NF-kappaB/c-fos signal transduction pathway. breviscapine 12-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-218 19383518-0 2009 Analgesic effect of milnacipran is associated with c-Fos expression in the anterior cingulate cortex in the rat neuropathic pain model. Milnacipran 20-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 18849175-11 2009 Furthermore, minocycline decreased the expression of noxious-stimulation-induced c-Fos, suggesting an effect on evoked neuronal activity. Minocycline 13-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-86 19470384-5 2009 During the postadolescent period (10-14 weeks postnatal), immunohistochemical experiments were carried out to investigate c-Fos expression following the administration of R-(+)-8-hydroxy-2-(di-n-propylamino) tetralin (8-OH-DPAT), a 5-HT(1A) receptor agonist. r-(+)-8-hydroxy-2-(di-n-propylamino) tetralin 171-216 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 19470384-6 2009 In the 3wFS group, the 8-OH-DPAT-induced c-Fos expression in the medial prefrontal cortex was significantly attenuated compared to that in the non-FS control group. 8-Hydroxy-2-(di-n-propylamino)tetralin 23-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 19470384-6 2009 In the 3wFS group, the 8-OH-DPAT-induced c-Fos expression in the medial prefrontal cortex was significantly attenuated compared to that in the non-FS control group. phenylalanylserine 9-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 19383518-5 2009 The increased level of c-Fos expression in the anterior cingulate cortex (ACC) induced by noxious mechanical stimulation was significantly inhibited by the continuous administration of milnacipran, indicating that milnacipran might cause a functional modification in the nociceptive processing in the ACC. Milnacipran 185-196 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 19383518-5 2009 The increased level of c-Fos expression in the anterior cingulate cortex (ACC) induced by noxious mechanical stimulation was significantly inhibited by the continuous administration of milnacipran, indicating that milnacipran might cause a functional modification in the nociceptive processing in the ACC. Milnacipran 214-225 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 19383537-1 2009 These studies tested the hypothesis that c-fos, c-jun and AP-1 are early markers of platinum analogue-induced proximal tubule nephrotoxicity in primary rat proximal tubule (RPT) and human proximal tubule (HPT) cell cultures. Platinum 84-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 21155252-11 2009 CONCLUSION: Curcumin could attenuate the activation of p-ERK, p-CREB, c-fos in dorsal root ganglion to ameliorate the CCI-induced neuropathic pain. Curcumin 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 19467261-0 2009 Cocaine-induced Fos expression is detectable in the frontal cortex and striatum of rats under isoflurane but not alpha-chloralose anesthesia: implications for FMRI. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 19467261-0 2009 Cocaine-induced Fos expression is detectable in the frontal cortex and striatum of rats under isoflurane but not alpha-chloralose anesthesia: implications for FMRI. Isoflurane 94-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-19 19467261-1 2009 The ability of intravenous cocaine to induce Fos protein expression in anesthetized rats was tested. Cocaine 27-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 19467261-13 2009 In both experiments, the frontal cortex and striatum of the cocaine-treated nonanesthetized rats expressed Fos in greater amounts than the saline-treated nonanesthetized rats, as expected. Cocaine 60-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-110 19467261-14 2009 The alpha-chloralose treatment prevented cocaine-induced Fos expression across all eight subregions of the striatum and frontal cortex that were examined. Cocaine 41-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 19467261-15 2009 In contrast, isoflurane only partially attenuated Fos expression in the orbital and Cg2 subregions of frontal cortex. Isoflurane 13-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 19641116-6 2009 L-type VDCC antagonism by verapamil in perirhinal cortex during memory acquisition disrupted the normal pattern of differential Fos expression, so paralleling the antagonist-induced memory impairment. Verapamil 26-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 128-131 19641121-7 2009 Furthermore, mGlu5 receptor antagonism (10 mg/kg MPEP) significantly inhibited neuronal activity in the nucleus accumbens core as levels of the transcription factor c-Fos were significantly reduced. 6-methyl-2-(phenylethynyl)pyridine 49-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 19464270-8 2009 An increased c-fos immunoreactivity in kindled rats with CD exposed to convulsive PTZ challenge was also observed with LEV pretreatment. Pentylenetetrazole 82-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 19464270-8 2009 An increased c-fos immunoreactivity in kindled rats with CD exposed to convulsive PTZ challenge was also observed with LEV pretreatment. Levetiracetam 119-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 19633138-5 2009 Presentation of the CS in the extinction context yielded low levels of conditioned freezing and induced c-Fos expression in the infralimbic division of the medial prefrontal cortex, the intercalated nuclei of the amygdala, and the dentate gyrus (DG). Cesium 20-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-109 19633138-6 2009 In contrast, presentation of the CS outside of the extinction context yielded high levels of conditioned freezing and induced c-Fos expression in the prelimbic division of the medial prefrontal cortex, the lateral and basolateral nuclei of the amygdala, and the medial division of the central nucleus of the amygdala. Cesium 33-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 19633138-7 2009 Hippocampal areas CA1 and CA3 exhibited c-Fos expression when the CS was presented in either context. Cesium 66-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 19374938-0 2009 Striatal dopamine and glutamate receptors modulate methamphetamine-induced cortical Fos expression. Dopamine 9-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 19374938-0 2009 Striatal dopamine and glutamate receptors modulate methamphetamine-induced cortical Fos expression. Methamphetamine 51-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 19374938-6 2009 The nuclear expression of Fos, the protein product of the IEG c-fos, was quantified in both the striatum and the cortex of animals receiving intrastriatal dopamine or glutamate antagonist administration. Dopamine 155-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 19374938-6 2009 The nuclear expression of Fos, the protein product of the IEG c-fos, was quantified in both the striatum and the cortex of animals receiving intrastriatal dopamine or glutamate antagonist administration. Glutamic Acid 167-176 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 19241444-5 2009 Hypoxia-induced MMP-13 overexpression was antagonized by transfection with antisense oligodeoxynucleotides (AS-ODN) of c-Fos or c-Jun. Oligodeoxyribonucleotides 85-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 19225867-4 2009 Furthermore, mRNA levels for c-Fos and c-Jun, unlike other transcriptional factors, were increased by both NE and phenylephrine, a specific alpha(1)-AR agonist. Phenylephrine 114-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 19225867-5 2009 Increases in c-Fos and c-Jun gene expression due to NE were attenuated by both prazosin and a PLC inhibitor, U73122. Prazosin 79-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 19225867-5 2009 Increases in c-Fos and c-Jun gene expression due to NE were attenuated by both prazosin and a PLC inhibitor, U73122. 1-(6-((3-methoxyestra-1,3,5(10)-trien-17-yl)amino)hexyl)-1H-pyrrole-2,5-dione 109-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 19225867-6 2009 Activation of protein kinase C (PKC) with phorbol myristate acetate increased c-Fos and c-Jun mRNA, whereas inhibition of PKC with bisindolylmaleimide as well as inhibition of extracellular signal-regulated kinases (ERK) 1/2 with PD98059 abolished the NE-induced increase in c-Fos and c-Jun gene expression. Tetradecanoylphorbol Acetate 42-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 19295452-8 2009 Glucose increased c-fos and decreased c-jun expressions. Glucose 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 19295452-11 2009 CONCLUSIONS: Our results suggest that short-term changes of mitogen-activated protein kinase and AKT signaling pathways and c-fos and c-jun expressions caused by glucose are abolished by palmitate through phosphatidylinositol 3-kinase inhibition via ceramide synthesis. Glucose 162-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 19295452-11 2009 CONCLUSIONS: Our results suggest that short-term changes of mitogen-activated protein kinase and AKT signaling pathways and c-fos and c-jun expressions caused by glucose are abolished by palmitate through phosphatidylinositol 3-kinase inhibition via ceramide synthesis. Palmitates 187-196 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 19295452-11 2009 CONCLUSIONS: Our results suggest that short-term changes of mitogen-activated protein kinase and AKT signaling pathways and c-fos and c-jun expressions caused by glucose are abolished by palmitate through phosphatidylinositol 3-kinase inhibition via ceramide synthesis. Ceramides 250-258 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 19639751-9 2009 By comparison, microinjection of BIC into CNA prior to IS attenuated both the reduction in REM and Fos expression in LC to levels seen in non-shocked controls. Bicuculline 33-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 19460425-9 2009 Furthermore, in males and females, the c-Fos response of npEW-Ucn1 neurons upon restraint stress was blunted in animals with MS history, a phenomenon that was concomitant with dampening of the HPA corticosterone response in females but not in males. Corticosterone 197-211 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 19693978-10 2009 The greatest density of Fos-positive neurons was located in lamine I-II in Group I. Serum IL-6 levels were significantly elevated in Group I. Pretreatment with tramadol showed a dose-depended inhibitory effect on c-fos expression and serum IL-6 production,but not in Group T1. Tramadol 160-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 213-218 19693978-11 2009 Administration of tramadol postoperatively also suppressed the c-fos expression and serum IL-6 production as showed in PT10 but were weaker than those in Group T10. Tramadol 18-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 19693978-0 2009 [Tramadol inhibits c-fos expression in spinal cord dorsal horn and serum IL-6 levels induced by plantar incision in rats]. Tramadol 1-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 19693978-1 2009 OBJECTIVE: To investigate effect of tramadol on c-fos expression in spinal cord dorsal horn and serum IL-6 levels induced by plantar incision in rats. Tramadol 36-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 19693978-12 2009 CONCLUSION: Pretreatment with tramadol can produce dose-dependent inhibitory effect on c-fos expression in spinal cord dorsal horn and then suppress the inflammatory response to the trauma. Tramadol 30-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 19693978-10 2009 The greatest density of Fos-positive neurons was located in lamine I-II in Group I. Serum IL-6 levels were significantly elevated in Group I. Pretreatment with tramadol showed a dose-depended inhibitory effect on c-fos expression and serum IL-6 production,but not in Group T1. Tramadol 160-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 19777800-0 2009 Comparative study of c-Fos expression in rat dorsal vagal complex and nucleus ambiguus induced by different durations of restraint water-immersion stress. Water 131-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-26 19777800-6 2009 Interestingly, the most intense c-Fos expression was observed in the dorsal motor nucleus of the vagus (DMV) during the stress, followed by NA, nucleus of solitary tract (NTS) and area postrema (AP). (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 104-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 19777800-7 2009 The peak of c-Fos expression in caudal DMV appeared at 120 min of the stress, slower than that in rostral and intermediate DMV. (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 39-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 19429152-4 2009 Using immunohistochemistry, we found that chronic fluoxetine treatment caused an increase in the expression of the early expression gene c-fos. Fluoxetine 50-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 19359223-10 2009 The Fos count after systemic LPS was reduced to 99+/-30 following pretreatment with the cyclooxygenase inhibitor Naproxen (10 mg kg(-1); p>0.05 versus vehicle controls) and increased to 242+/-66 following the iNOS-inhibitor Aminoguanidine (15 mg kg(-1); p<0.01). Naproxen 113-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-7 19359223-10 2009 The Fos count after systemic LPS was reduced to 99+/-30 following pretreatment with the cyclooxygenase inhibitor Naproxen (10 mg kg(-1); p>0.05 versus vehicle controls) and increased to 242+/-66 following the iNOS-inhibitor Aminoguanidine (15 mg kg(-1); p<0.01). pimagedine 227-241 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-7 19362521-0 2009 Brain Fos expression induced by the chemotherapy agent cisplatin in the rat is partially dependent on an intact abdominal vagus. Cisplatin 55-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 6-9 19362521-4 2009 Cisplatin also stimulates Fos expression in the rat brain in areas known to play a role in emesis in other species, but it is not known whether vagal input is required for this CNS activation. Cisplatin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 19362521-5 2009 In the present study, rats were given abdominal vagotomy or sham operation to test the role of an intact vagus on cisplatin-induced Fos expression 6 h after injection with saline or cisplatin (6 mg/kg, ip). Cisplatin 114-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-135 19362521-6 2009 Cisplatin treatment produced Fos expression in the area postrema and multiple levels of the nucleus of the solitary tract (NTS) of sham-operated rats. Cisplatin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 19362521-7 2009 Vagotomy reduced cisplatin-induced Fos expression in the caudal and middle levels of the NTS and central amygdala. Cisplatin 17-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 19362521-9 2009 These results suggest that a defined portion of cisplatin-induced Fos expression is dependent on vagal input, with a majority of this response determined by either direct action of cisplatin or humoral factors on the CNS. Cisplatin 48-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 19490646-6 2009 Lafutidine reduced the HCl-evoked expression of c-Fos in the NTS and elevated the intragastric pH following intragastric administration of excess HCl. lafutidine 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 19490646-6 2009 Lafutidine reduced the HCl-evoked expression of c-Fos in the NTS and elevated the intragastric pH following intragastric administration of excess HCl. Hydrochloric Acid 23-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 19348787-5 2009 Following PDA stimulation, the number of Fos-like immunoreactive neurons increased in the caudal and intermediate parts of the nucleus of the solitary tract (NST), the area postrema (AP), the external lateral subnucleus of the parabrachial nucleus (PBN), the arcuate nucleus of the hypothalamus (Arc), and the central amygdaloid nucleus (CeA). 2,3-piperidinedicarboxylic acid 10-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 19462165-6 2009 A considerable numbers of c-Fos-immunoreactive (IR) cells were induced in the caudal and inter-medio-lateral center of superficial layers of the Vc (VcI/II; mean +/- SEM/section = 225.8 +/- 12.9) and magnocellular zone of the Vc (VcIII/IV; 67.1 +/- 4.7) 2 h after formalin injection into the lip. Formaldehyde 264-272 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 19462165-7 2009 Much smaller numbers of c-Fos-IR cells were induced in the rostral and dorso-medial one-fourth of the VcI/II (72.6 +/- 3.7) and VcIII/IV (55.6 +/- 6.6) after formalin injection into the tongue. Formaldehyde 158-166 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 19462165-8 2009 Following preadministration with systemic bicuculline or morphine, the formalin-induced c-Fos-IR cells were decreased more in the VcI/II when formalin was injected into the lip (VcI/II, 102.4 +/- 8.0; VcIII/IV, 32.8 +/- 1.4) than into the tongue (VcI/II, 49.5 +/- 8.1; VcIII/IV, 31.7 +/- 5.3). Bicuculline 42-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 19462165-8 2009 Following preadministration with systemic bicuculline or morphine, the formalin-induced c-Fos-IR cells were decreased more in the VcI/II when formalin was injected into the lip (VcI/II, 102.4 +/- 8.0; VcIII/IV, 32.8 +/- 1.4) than into the tongue (VcI/II, 49.5 +/- 8.1; VcIII/IV, 31.7 +/- 5.3). Morphine 57-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 19462165-8 2009 Following preadministration with systemic bicuculline or morphine, the formalin-induced c-Fos-IR cells were decreased more in the VcI/II when formalin was injected into the lip (VcI/II, 102.4 +/- 8.0; VcIII/IV, 32.8 +/- 1.4) than into the tongue (VcI/II, 49.5 +/- 8.1; VcIII/IV, 31.7 +/- 5.3). Formaldehyde 71-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 19462165-8 2009 Following preadministration with systemic bicuculline or morphine, the formalin-induced c-Fos-IR cells were decreased more in the VcI/II when formalin was injected into the lip (VcI/II, 102.4 +/- 8.0; VcIII/IV, 32.8 +/- 1.4) than into the tongue (VcI/II, 49.5 +/- 8.1; VcIII/IV, 31.7 +/- 5.3). Formaldehyde 142-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 19201768-7 2009 RESULTS: Either NaHS or capsaicin induced the expression of Fos protein in the superficial layers of the T8 and T9 spinal dorsal horn of rats or mice. sodium bisulfide 16-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 19201768-7 2009 RESULTS: Either NaHS or capsaicin induced the expression of Fos protein in the superficial layers of the T8 and T9 spinal dorsal horn of rats or mice. Capsaicin 24-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 19115412-6 2009 To establish the effects of 5-HT1AR stimulation on L-DOPA-induced c-fos and preprodynorphin (PPD) mRNA within the dopamine-depleted striatum, immunohistochemistry and real-time reverse transcription polymerase chain reaction, respectively, were used. Levodopa 51-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 19115412-9 2009 Striatal c-fos immunoreactivity and PPD mRNA ipsilateral to the lesion were strongly induced by L-DOPA, while +/-8-OH-DPAT suppressed these effects. Levodopa 96-102 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-14 18656400-9 2009 These drugs inhibited c-fos expression in the superficial layer of the spinal dorsal horn of segments L4-5 at 2h after formalin injection. Formaldehyde 119-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 19169671-0 2009 Drug context differently regulates cocaine versus heroin self-administration and cocaine- versus heroin-induced Fos mRNA expression in the rat. Cocaine 81-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 19169671-0 2009 Drug context differently regulates cocaine versus heroin self-administration and cocaine- versus heroin-induced Fos mRNA expression in the rat. Heroin 97-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 19169671-8 2009 RESULTS: We found that: (1) drug-taking context differentially modulates intravenous cocaine versus heroin self-administration; (2) very low doses of cocaine and heroin are sufficient to induce Fos mRNA expression in the posterior caudate; (3) drug-administration context differentially modulates cocaine- versus heroin-induced Fos mRNA expression. Cocaine 85-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 194-197 19169671-8 2009 RESULTS: We found that: (1) drug-taking context differentially modulates intravenous cocaine versus heroin self-administration; (2) very low doses of cocaine and heroin are sufficient to induce Fos mRNA expression in the posterior caudate; (3) drug-administration context differentially modulates cocaine- versus heroin-induced Fos mRNA expression. Cocaine 150-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 194-197 19169671-8 2009 RESULTS: We found that: (1) drug-taking context differentially modulates intravenous cocaine versus heroin self-administration; (2) very low doses of cocaine and heroin are sufficient to induce Fos mRNA expression in the posterior caudate; (3) drug-administration context differentially modulates cocaine- versus heroin-induced Fos mRNA expression. Cocaine 150-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 328-331 19169671-8 2009 RESULTS: We found that: (1) drug-taking context differentially modulates intravenous cocaine versus heroin self-administration; (2) very low doses of cocaine and heroin are sufficient to induce Fos mRNA expression in the posterior caudate; (3) drug-administration context differentially modulates cocaine- versus heroin-induced Fos mRNA expression. Cocaine 150-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 194-197 19169671-8 2009 RESULTS: We found that: (1) drug-taking context differentially modulates intravenous cocaine versus heroin self-administration; (2) very low doses of cocaine and heroin are sufficient to induce Fos mRNA expression in the posterior caudate; (3) drug-administration context differentially modulates cocaine- versus heroin-induced Fos mRNA expression. Cocaine 150-157 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 328-331 19474332-10 2009 In the MPTA itself FOS expression was suppressed during systemic anesthesia. mpta 7-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 19243923-2 2009 Systemic administration of BU224 or harmane to naive rats increased Fos-like immunoreactivity (FLI) in the hypothalamic paraventricular nucleus (PVN), hippocampal dentate gyrus (DG), central and medial nuclei of the amygdala (CeA, MeA) and the locus coeruleus (LC). BU 224 27-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 19243923-2 2009 Systemic administration of BU224 or harmane to naive rats increased Fos-like immunoreactivity (FLI) in the hypothalamic paraventricular nucleus (PVN), hippocampal dentate gyrus (DG), central and medial nuclei of the amygdala (CeA, MeA) and the locus coeruleus (LC). harman 36-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-71 19409080-7 2009 Suppression of SND genesis by tetrodotoxin (TTX) or mecamylamine (MECA, nicotinic receptor blocker) almost abolished c-Fos expression in dorsal laminae, but only mildly affected c-Fos expression in the SPNs. Tetrodotoxin 30-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 19409080-7 2009 Suppression of SND genesis by tetrodotoxin (TTX) or mecamylamine (MECA, nicotinic receptor blocker) almost abolished c-Fos expression in dorsal laminae, but only mildly affected c-Fos expression in the SPNs. Tetrodotoxin 44-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 19409080-7 2009 Suppression of SND genesis by tetrodotoxin (TTX) or mecamylamine (MECA, nicotinic receptor blocker) almost abolished c-Fos expression in dorsal laminae, but only mildly affected c-Fos expression in the SPNs. Mecamylamine 52-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 19409080-7 2009 Suppression of SND genesis by tetrodotoxin (TTX) or mecamylamine (MECA, nicotinic receptor blocker) almost abolished c-Fos expression in dorsal laminae, but only mildly affected c-Fos expression in the SPNs. Mecamylamine 52-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 178-183 19416632-8 2009 Treating healthy rats with NO2-Arg-Trim resulted in a dose-dependent attenuation of CRD-induced nociception and in an inhibition of CRD-induced overexpression of spinal cFOS mRNA. Nitrogen Dioxide 27-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-173 19115403-0 2009 The action of pulse-modulated GSM radiation increases regional changes in brain activity and c-Fos expression in cortical and subcortical areas in a rat model of picrotoxin-induced seizure proneness. Picrotoxin 162-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 19115403-2 2009 In the present study, GSM-exposed picrotoxin-pretreated rats showed differences in clinical and EEG signs, and in c-Fos expression in the brain, with respect to picrotoxin-treated rats exposed to an equivalent dose of unmodulated radiation. Picrotoxin 34-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 19115403-4 2009 The most marked effects of GSM radiation on c-Fos expression in picrotoxin-treated rats were observed in limbic structures, olfactory cortex areas and subcortical areas, the dentate gyrus, and the central lateral nucleus of the thalamic intralaminar nucleus group. Picrotoxin 64-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 19371588-8 2009 alone did not change the architecture of pentobarbital sleep and pentobarbital-induced c-Fos expression in the VLPO and the TMN, but co-administration of them significantly increased both total pentobarbital sleep and SWS, whereas decreased REM sleep, with increasing c-Fos expression in the VLPO and concomitantly decreasing c-Fos expression in the TMN. Pentobarbital 65-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 19371588-8 2009 alone did not change the architecture of pentobarbital sleep and pentobarbital-induced c-Fos expression in the VLPO and the TMN, but co-administration of them significantly increased both total pentobarbital sleep and SWS, whereas decreased REM sleep, with increasing c-Fos expression in the VLPO and concomitantly decreasing c-Fos expression in the TMN. Pentobarbital 65-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 19249327-0 2009 Armodafinil promotes wakefulness and activates Fos in rat brain. Modafinil 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 19249327-1 2009 Modafinil increases waking and labeling of Fos, a marker of neuronal activation. Modafinil 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-46 19249327-6 2009 Armodafinil had differential effects in increasing neuronal Fos immunolabeling 2 h after administration. Modafinil 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 19249327-7 2009 Armodafinil at 100 mg/kg increased numbers of Fos-labeled neurons in striatum and anterior cingulate cortex, without affecting nucleus accumbens. Modafinil 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 19249327-8 2009 Armodafinil at 30 mg/kg only increased numbers of light Fos-labeled neurons in the anterior cingulate cortex. Modafinil 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-59 19249327-9 2009 In brainstem arousal centers, 100 mg/kg armodafinil increased numbers of Fos-labeled neurons in the tuberomammillary nucleus, pedunculopontine tegmentum, laterodorsal tegmentum, locus coeruleus, and dorsal raphe nucleus. Modafinil 40-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-76 19249327-10 2009 Fos activation of these brainstem arousal centers, as well as of the cortex and striatum, is consistent with the observed arousal effects of armodafinil. Modafinil 141-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 19416632-8 2009 Treating healthy rats with NO2-Arg-Trim resulted in a dose-dependent attenuation of CRD-induced nociception and in an inhibition of CRD-induced overexpression of spinal cFOS mRNA. Arginine 31-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-173 19416632-10 2009 In this setting,NO2-Arg-Trim but not trimebutine, significantly down-regulated the spinal cFOS mRNA expression and increased blood concentrations of NO2 +NO3. Nitroarginine 16-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-94 19390822-0 2009 Lipoxygenase inhibitors suppressed carrageenan-induced Fos-expression and inflammatory pain responses in the rat. Carrageenan 35-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-58 19390822-4 2009 Intradermally injected carrageenan caused elevated number of cells exhibiting Fos-like immunoreactivity (Fos-LI) in the spinal dorsal horn, and decreased the thermal and mechanical threshold in Hargreaves and von Frey tests. Carrageenan 23-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 19390822-4 2009 Intradermally injected carrageenan caused elevated number of cells exhibiting Fos-like immunoreactivity (Fos-LI) in the spinal dorsal horn, and decreased the thermal and mechanical threshold in Hargreaves and von Frey tests. Carrageenan 23-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-108 19390822-5 2009 Pretreatment with an inhibitor of phospholipase A2, that generates the LO substrate, prior to the carrageenan injection significantly reduced the number of Fos-(+) cells. Carrageenan 98-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 156-159 19390822-7 2009 Moreover, the LO inhibitors suppressed carrageenan-induced thermal and mechanical hyperalgesic behaviors, which inidcates that the changes in Fos expression correlates with those in the nociceptive behaviors in the inflamed rats. Carrageenan 39-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-145 19390822-9 2009 Overall, our results from the Fos-LI and behavior tests suggest that LO products released from inflamed tissues contribute to nociception during carrageenan-induced inflammation, indicating that the LO pathway is a possible target for modulating inflammatory pain. Carrageenan 145-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 19232381-1 2009 Expression of c-fos in the medial geniculate body (MGB) and the inferior colliculus (IC) in response to bicuculline-induced corticofugal activation was examined in rats at different time points after bilateral cochlear ablation (4 h-30 days). Bicuculline 104-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 19232381-8 2009 After bilateral cochlear ablation, significant increases in Fos-positive neurons were detected unilaterally in all IC subnuclei, ipsilateral to the bicuculline injection. Bicuculline 148-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 19248773-9 2009 Exercise decreased the severity of or eliminated seizure behaviors and hippocampal c-fos expression induced by kainic acid. Kainic Acid 111-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 19429022-7 2009 Aminoguanidine reduced c-fos expression in the AP and NTS at 6h after CLP, but showed an opposite effect at 24h, with an increase in the AP, NTS, and also in the VLM. pimagedine 0-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 19548582-0 2009 [Effects of lanthanum on memory and expression of c-fos mRNA and c-Fos protein of cerebral cortex in rats]. Lanthanum 12-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 19548582-0 2009 [Effects of lanthanum on memory and expression of c-fos mRNA and c-Fos protein of cerebral cortex in rats]. Lanthanum 12-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 19548582-7 2009 The expression levels of c-fos mRNA and c-Fos protein in cerebral cortex of low dose lanthanum group were lower significantly than control values. Lanthanum 85-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 19548582-7 2009 The expression levels of c-fos mRNA and c-Fos protein in cerebral cortex of low dose lanthanum group were lower significantly than control values. Lanthanum 85-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 19548582-8 2009 As to middle dose lanthanum group, the expression levels of c-fos mRNA in cerebral cortex were lower significantly than control values, and c-Fos protein expression were lower significantly than control and low dose lanthanum group. Lanthanum 18-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 19548582-9 2009 Both c-fos mRNA and c-Fos protein expression in cerebral cortex of high dose lanthanum group decreased significantly as compared with control and low dose lanthanum group. Lanthanum 77-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 5-10 19548582-9 2009 Both c-fos mRNA and c-Fos protein expression in cerebral cortex of high dose lanthanum group decreased significantly as compared with control and low dose lanthanum group. Lanthanum 77-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 19548582-9 2009 Both c-fos mRNA and c-Fos protein expression in cerebral cortex of high dose lanthanum group decreased significantly as compared with control and low dose lanthanum group. Lanthanum 155-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 5-10 19225146-0 2009 Chemotherapy agent cisplatin induces 48-h Fos expression in the brain of a vomiting species, the house musk shrew (Suncus murinus). Cisplatin 19-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 19225146-4 2009 Our prior data show that brain Fos expression is increased for at least 48 h after cisplatin treatment in the rat, a nonvomiting species. Cisplatin 83-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 19225146-6 2009 Compared with saline injection, cisplatin treatment (30 mg/kg ip) induced Fos expression in hindbrain areas known to play a role in the generation of emesis, the dorsal motor nucleus (DMN), the area postrema, and the nucleus of the solitary tract (NTS), for up to 48 h. Cisplatin also stimulated Fos expression in the parabrachial nucleus (PBN) of the midbrain and the central nucleus of the amygdala (CeA) for at least 48 h after treatment. Cisplatin 32-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 19225146-6 2009 Compared with saline injection, cisplatin treatment (30 mg/kg ip) induced Fos expression in hindbrain areas known to play a role in the generation of emesis, the dorsal motor nucleus (DMN), the area postrema, and the nucleus of the solitary tract (NTS), for up to 48 h. Cisplatin also stimulated Fos expression in the parabrachial nucleus (PBN) of the midbrain and the central nucleus of the amygdala (CeA) for at least 48 h after treatment. Cisplatin 32-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 296-299 19225146-7 2009 When animals were pretreated with the antiemetic palonosetron, a long-term serotonin type 3 (5-HT(3)) receptor antagonist, cisplatin-induced Fos expression was significantly attenuated in the NTS, DMN, and CeA at 6 h but not at 48 h. These results indicate that cisplatin activates a neural system that includes the dorsal vagal complex and forebrain in the musk shrew, which is partially suppressed by a 5-HT(3) receptor antagonist. Palonosetron 49-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 19225146-7 2009 When animals were pretreated with the antiemetic palonosetron, a long-term serotonin type 3 (5-HT(3)) receptor antagonist, cisplatin-induced Fos expression was significantly attenuated in the NTS, DMN, and CeA at 6 h but not at 48 h. These results indicate that cisplatin activates a neural system that includes the dorsal vagal complex and forebrain in the musk shrew, which is partially suppressed by a 5-HT(3) receptor antagonist. Serotonin 75-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 19225146-7 2009 When animals were pretreated with the antiemetic palonosetron, a long-term serotonin type 3 (5-HT(3)) receptor antagonist, cisplatin-induced Fos expression was significantly attenuated in the NTS, DMN, and CeA at 6 h but not at 48 h. These results indicate that cisplatin activates a neural system that includes the dorsal vagal complex and forebrain in the musk shrew, which is partially suppressed by a 5-HT(3) receptor antagonist. Cisplatin 123-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 19225146-7 2009 When animals were pretreated with the antiemetic palonosetron, a long-term serotonin type 3 (5-HT(3)) receptor antagonist, cisplatin-induced Fos expression was significantly attenuated in the NTS, DMN, and CeA at 6 h but not at 48 h. These results indicate that cisplatin activates a neural system that includes the dorsal vagal complex and forebrain in the musk shrew, which is partially suppressed by a 5-HT(3) receptor antagonist. 2,3-dimethylnaphthalene 197-200 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 19225146-7 2009 When animals were pretreated with the antiemetic palonosetron, a long-term serotonin type 3 (5-HT(3)) receptor antagonist, cisplatin-induced Fos expression was significantly attenuated in the NTS, DMN, and CeA at 6 h but not at 48 h. These results indicate that cisplatin activates a neural system that includes the dorsal vagal complex and forebrain in the musk shrew, which is partially suppressed by a 5-HT(3) receptor antagonist. Cisplatin 262-271 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 18723099-8 2009 Fos+/TH+/HRP+/GFAP+ and Fos+/VP+/HRP+/GFAP+ quadruplicate labeled N-ASC could be found in the MVZ, PVN and SON, respectively. n-asc 66-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 18723099-8 2009 Fos+/TH+/HRP+/GFAP+ and Fos+/VP+/HRP+/GFAP+ quadruplicate labeled N-ASC could be found in the MVZ, PVN and SON, respectively. n-asc 66-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 19220411-7 2009 In Proechimys, almost all reticular nucleus neurons were Fos-positive at 24 h. DISCUSSION: At variance with laboratory rats, pilocarpine-induced protracted seizures elicit in Proechimys limited neuronal death, and marked and long-lasting Fos induction in excitatory and inhibitory cortical and thalamic cell subsets. Pilocarpine 125-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 19220411-7 2009 In Proechimys, almost all reticular nucleus neurons were Fos-positive at 24 h. DISCUSSION: At variance with laboratory rats, pilocarpine-induced protracted seizures elicit in Proechimys limited neuronal death, and marked and long-lasting Fos induction in excitatory and inhibitory cortical and thalamic cell subsets. Pilocarpine 125-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 238-241 19172228-0 2009 Melatonin treatment decreases c-fos expression in a headache model induced by capsaicin. Melatonin 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 19172228-0 2009 Melatonin treatment decreases c-fos expression in a headache model induced by capsaicin. Capsaicin 78-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 19172228-4 2009 On the other hand, pinealectomized rats, which received capsaicin, presented the highest number of c-fos-positive cells. Capsaicin 56-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 18953528-0 2009 The impact of early environmental rearing condition on the discriminative stimulus effects and Fos expression induced by cocaine in adult male and female rats. Cocaine 121-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-98 18953528-2 2009 OBJECTIVES: This study assessed if MS affected the stimulus and Fos-inducing effects of cocaine. Cocaine 88-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 19116947-0 2009 Differential regulation of prodynophin, c-fos, and serotonin transporter mRNA following withdrawal from a chronic, escalating dose regimen of D-amphetamine. Dextroamphetamine 142-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 19116947-4 2009 This study examined the effects of chronic, escalating doses of D-AMPH followed by 24 h of withdrawal on the expression of prodynorphin (PD) and c-fos mRNA in limbic regions of the brain, caudate putamen (CPu), and brainstem and SERT mRNA expression in the dorsal raphe nucleus (DRN). Dextroamphetamine 64-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-150 19201780-8 2009 Interestingly, MetaCore pathway analysis grouped the majority of these proteins around two principal nodes (c-fos and c-myc) suggesting that they may participate in the transcriptional activation of key pathways in TCDD-driven inhibition of osteoblast differentiation. Polychlorinated Dibenzodioxins 215-219 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 19136019-7 2009 NTX administered intra-CeA reduced c-Fos-immunoreactivity (IR) in nucleus accumbens neurons significantly compared to the intra-PVN MTII treated animals, animals co-injected intra-PVN with MTII and intra-CeA with NTX animals, and control animals. Naltrexone 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 19136019-9 2009 Intra-CeA NTX co-injected with intra-PVN MTII induced c-Fos-IR significantly in PVN neurons relative to control and intra-CeA NTX animals. Naltrexone 10-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 19084559-0 2009 Expression of c-fos mRNA in the basal ganglia associated with contingent tolerance to amphetamine-induced hypophagia. Amphetamine 86-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 19084559-6 2009 Following an acute injection of amphetamine, both of these groups had higher levels of c-fos mRNA than saline-treated controls throughout the striatum, in the nucleus accumbens core, the ventral pallidum and layers V-VI of the motor cortex. Amphetamine 32-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 19154780-8 2009 LPS both histamine- and dry eye-evoked Fos was increased at the Vi/Vc transition, while smaller effects were seen at other regions. Histamine 9-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 19154780-10 2009 Enhancement of Fos at the Vi/Vc region after MO, histamine and dry eye conditions supports the hypothesis that this region integrates innocuous as well as noxious sensory information, while more caudal portions of Vc process mainly nociceptive signals from the eye. Histamine 49-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-18 19174180-3 2009 Initial immunohistochemical measurements of the marker of neural activation, Fos, confirmed that in halothane-anesthetized rats fenfluramine (10 mg/kg i.v.) Fenfluramine 128-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-80 19174180-6 2009 Fenfluramine-induced Fos was demonstrated in numerous glutamatergic pyramidal neurons (Fos/excitatory amino acid carrier 1 (EAAC1) co-labeled), but also a small number of GABA interneurons (Fos/glutamic acid decarboxylase (GAD)(67) colabeled). Fenfluramine 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 19174180-6 2009 Fenfluramine-induced Fos was demonstrated in numerous glutamatergic pyramidal neurons (Fos/excitatory amino acid carrier 1 (EAAC1) co-labeled), but also a small number of GABA interneurons (Fos/glutamic acid decarboxylase (GAD)(67) colabeled). Fenfluramine 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 19174180-6 2009 Fenfluramine-induced Fos was demonstrated in numerous glutamatergic pyramidal neurons (Fos/excitatory amino acid carrier 1 (EAAC1) co-labeled), but also a small number of GABA interneurons (Fos/glutamic acid decarboxylase (GAD)(67) colabeled). Fenfluramine 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 19285017-2 2009 In our previous studies, we found that in addition to SRE, CRE is required for maximal c-fos promoter activation triggered by lysophosphatidic acid (LPA). lysophosphatidic acid 126-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 19285017-2 2009 In our previous studies, we found that in addition to SRE, CRE is required for maximal c-fos promoter activation triggered by lysophosphatidic acid (LPA). lysophosphatidic acid 149-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 19285017-7 2009 Moreover, levels of c-fos mRNA and cyclin D1 protein were increased via LPA-induced CREB phosphorylation. lysophosphatidic acid 72-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 19120051-14 2009 Clonidine (60 microg/kg) or ethanol (1 g/kg) alone increased, but their combination decreased, c-Fos levels in LC, while inconsistent c-Fos responses were observed in cerebellum. Clonidine 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 19120051-14 2009 Clonidine (60 microg/kg) or ethanol (1 g/kg) alone increased, but their combination decreased, c-Fos levels in LC, while inconsistent c-Fos responses were observed in cerebellum. Ethanol 28-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 19254279-0 2009 Anti-apoptotic effects of protein kinase C-delta and c-fos in cisplatin-treated thyroid cells. Cisplatin 62-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-58 19254279-2 2009 The studies described herein examined whether c-fos was associated with cisplatin resistance and the signalling link between c-fos and PKC-delta/ERK. Cisplatin 72-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 19254279-4 2009 KEY RESULTS: Cisplatin provokes the induction of c-fos and the activation of conventional PKC-beta, and novel PKC-delta and -epsilon. Cisplatin 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 19254279-5 2009 The cisplatin-provoked c-fos induction was decreased by Go6976, a PKC-beta inhibitor; by siRNA for PKC-delta- but not that for PKC-epsilon or by PD98059, a mitogen-activated protein kinase/ERK kinase inhibitor. Cisplatin 4-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 19254279-5 2009 The cisplatin-provoked c-fos induction was decreased by Go6976, a PKC-beta inhibitor; by siRNA for PKC-delta- but not that for PKC-epsilon or by PD98059, a mitogen-activated protein kinase/ERK kinase inhibitor. Go 6976 56-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 19254279-5 2009 The cisplatin-provoked c-fos induction was decreased by Go6976, a PKC-beta inhibitor; by siRNA for PKC-delta- but not that for PKC-epsilon or by PD98059, a mitogen-activated protein kinase/ERK kinase inhibitor. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 145-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-28 19254279-6 2009 Expression of c-fos was abolished by GF109203X, an inhibitor of all PKC isoforms, or by PD98059 plus Go6976 or by PKC-delta-siRNA plus Go6976. bisindolylmaleimide I 37-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 19254279-6 2009 Expression of c-fos was abolished by GF109203X, an inhibitor of all PKC isoforms, or by PD98059 plus Go6976 or by PKC-delta-siRNA plus Go6976. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 88-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 19254279-6 2009 Expression of c-fos was abolished by GF109203X, an inhibitor of all PKC isoforms, or by PD98059 plus Go6976 or by PKC-delta-siRNA plus Go6976. Go 6976 101-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 19254279-6 2009 Expression of c-fos was abolished by GF109203X, an inhibitor of all PKC isoforms, or by PD98059 plus Go6976 or by PKC-delta-siRNA plus Go6976. Go 6976 135-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 19254279-7 2009 When c-fos expression was blocked by siRNA, cisplatin cytotoxicity was strongly enhanced with increased caspase-3 activation. Cisplatin 44-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 5-10 19074588-8 2009 In comparison with saline treatment, LPS administration induced lower food intake and increased plasma ACTH and corticosterone levels, as well as an increase in Fos-CRF and Fos-alpha-MSH double-labelled neurons in vehicle-pretreated rats. lps 37-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-164 19074588-8 2009 In comparison with saline treatment, LPS administration induced lower food intake and increased plasma ACTH and corticosterone levels, as well as an increase in Fos-CRF and Fos-alpha-MSH double-labelled neurons in vehicle-pretreated rats. lps 37-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 173-176 19074588-9 2009 In contrast, indomethacin treatment partly reversed the hypophagic effect, blunted the hormonal increase and blocked the Fos-CRF and Fos-alpha-MSH hypothalamic double labelling increase in response to the LPS stimulus. Indomethacin 13-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-124 19074588-9 2009 In contrast, indomethacin treatment partly reversed the hypophagic effect, blunted the hormonal increase and blocked the Fos-CRF and Fos-alpha-MSH hypothalamic double labelling increase in response to the LPS stimulus. Indomethacin 13-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-136 19074588-9 2009 In contrast, indomethacin treatment partly reversed the hypophagic effect, blunted the hormonal increase and blocked the Fos-CRF and Fos-alpha-MSH hypothalamic double labelling increase in response to the LPS stimulus. lps 205-208 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-124 19074588-9 2009 In contrast, indomethacin treatment partly reversed the hypophagic effect, blunted the hormonal increase and blocked the Fos-CRF and Fos-alpha-MSH hypothalamic double labelling increase in response to the LPS stimulus. lps 205-208 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-136 19705550-0 2009 Induction of Fos proteins in regions of the nucleus accumbens and ventrolateral striatum correlates with catalepsy and stereotypic behaviours induced by morphine. Morphine 153-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 19020866-0 2009 Persistence of one-trial cocaine-induced behavioral sensitization in young rats: regional differences in Fos immunoreactivity. Cocaine 25-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-108 19020866-8 2009 When assessed after three abstinence days (i.e., on PD 22), acute treatment with cocaine increased Fos-IR in various brain regions, but sensitized responding was associated with elevated Fos expression in only the caudate-putamen (CP) and prefrontal cortex (PFC). Cocaine 81-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 19020866-8 2009 When assessed after three abstinence days (i.e., on PD 22), acute treatment with cocaine increased Fos-IR in various brain regions, but sensitized responding was associated with elevated Fos expression in only the caudate-putamen (CP) and prefrontal cortex (PFC). Cocaine 81-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 187-190 19245875-5 2009 The purpose of the present study was to correlate cocaine-induced locomotor activity with neuronal activation in subregions of the striatum and cortex by acute cocaine in young adolescent (postnatal (PN) 28) and adult (PN 65) male rats by measuring the induction of the plasticity-associated immediate early genes (IEGs) c-fos and zif268 using in situ hybridization. Cocaine 50-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 321-326 19245875-7 2009 Low dose cocaine induced more locomotor activity and striatal c-fos expression in adolescents than adults whereas high dose cocaine induced more locomotor activity, striatal c-fos, and striatal zif268 expression in adults. Cocaine 9-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-67 19245875-7 2009 Low dose cocaine induced more locomotor activity and striatal c-fos expression in adolescents than adults whereas high dose cocaine induced more locomotor activity, striatal c-fos, and striatal zif268 expression in adults. Cocaine 124-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 174-179 19248818-10 2009 In rats, exposure to ethanol, psychostimulant and restraint stress all induced pIIIu Fos immunoreactivity compared to saline-injected controls. Ethanol 21-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 19248818-11 2009 In both mice and rats, ethanol- and cocaine-induced Fos immunoreactivity occurred exclusively in urocortin 1-positive, but not in tyrosine hydroxylase-positive, cells. Ethanol 23-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 19705550-5 2009 We examined the effect of intermittent morphine exposure on the distribution of Fos proteins in the basal ganglia following a subsequent morphine challenge administered after a period of drug abstinence. Morphine 39-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 19248818-11 2009 In both mice and rats, ethanol- and cocaine-induced Fos immunoreactivity occurred exclusively in urocortin 1-positive, but not in tyrosine hydroxylase-positive, cells. Cocaine 36-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 19705550-5 2009 We examined the effect of intermittent morphine exposure on the distribution of Fos proteins in the basal ganglia following a subsequent morphine challenge administered after a period of drug abstinence. Morphine 137-145 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 19705550-6 2009 We found that such exposures increased c-Fos induced by a morphine challenge in accumbens core regions that were immunoreactive for the micro-opioid receptor, and this correlated with the frequency of stereotypic behaviours displayed by the rats. Morphine 58-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 19705550-7 2009 We also found that a history of morphine exposures increased c-Fos in the ventrolateral striatum in response to a morphine challenge following 14 d but not 24 h of drug abstinence. Morphine 32-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 19705550-7 2009 We also found that a history of morphine exposures increased c-Fos in the ventrolateral striatum in response to a morphine challenge following 14 d but not 24 h of drug abstinence. Morphine 114-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 19118538-0 2009 NMDA preconditioning and neuroprotection in vivo: delayed onset of kainic acid-induced neurodegeneration and c-Fos attenuation in CA3a neurons. N-Methylaspartate 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 19118538-4 2009 In this study we attempted to prevent KA-induced damage in CA3 neurons with NMDA preconditioning, which produced a marked expression of c-fos in the hippocampus. N-Methylaspartate 76-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 19118538-9 2009 Quantitative evaluation of c-Fos-labeled cells showed significantly less c-Fos in CA3a than in neighboring CA3b and CA2 from 1 to 4 h after KA alone. ca3a 82-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 19118538-9 2009 Quantitative evaluation of c-Fos-labeled cells showed significantly less c-Fos in CA3a than in neighboring CA3b and CA2 from 1 to 4 h after KA alone. ca3a 82-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 19118538-9 2009 Quantitative evaluation of c-Fos-labeled cells showed significantly less c-Fos in CA3a than in neighboring CA3b and CA2 from 1 to 4 h after KA alone. ca3b 107-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 19118538-11 2009 NMDA preconditioning elevated CA3a c-Fos expression and at 1 and 2 h exceeded markedly that after KA alone. N-Methylaspartate 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 19118538-12 2009 However, at 4 h after KA, NMDA-preconditioned c-Fos induction in CA3a diminished to the same level as that seen after KA alone. N-Methylaspartate 26-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 19118538-12 2009 However, at 4 h after KA, NMDA-preconditioned c-Fos induction in CA3a diminished to the same level as that seen after KA alone. ca3a 65-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 19118538-14 2009 While NMDA-induced c-Fos expression in CA3a could be blocked by MK-801 completely, MK-801 and CNQX were both without significant effect on KA-induced c-Fos expression and neuronal damage. N-Methylaspartate 6-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 19118538-14 2009 While NMDA-induced c-Fos expression in CA3a could be blocked by MK-801 completely, MK-801 and CNQX were both without significant effect on KA-induced c-Fos expression and neuronal damage. Dizocilpine Maleate 64-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 19210763-0 2009 The signal transduction pathway of PKC/NF-kappa B/c-fos may be involved in the influence of high glucose on the cardiomyocytes of neonatal rats. Glucose 97-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-55 18976678-0 2009 Masculinization induced by neonatal exposure to PGE(2) or estradiol alters c-fos induction by estrous odors in adult rats. Prostaglandins E 48-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 18976678-0 2009 Masculinization induced by neonatal exposure to PGE(2) or estradiol alters c-fos induction by estrous odors in adult rats. Estradiol 58-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 19210763-2 2009 OBJECTIVES: To study the influence of high glucose on the structure, function and signal transduction pathway of PKC (Protein Kinase C)/NF-kappaB(Nuclear factor-kappaB)/c-fos in cultured cardiomyocytes. Glucose 43-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 169-174 19210763-5 2009 RESULTS: Cardiomyocytes cultured in high glucose level, but not iso-osmotic mannital, showed an increased pulsatile frequency and higher cellular volumes consistent with the increased glucose levels, and also higher expression of PKC-alpha, PKC-beta2, p-PKC-alpha, p-PKC-beta2, NF-kappaB, p-NF-kappaB, TNF-alpha and c-fos. Glucose 41-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 316-321 19210763-7 2009 CONCLUSION: High glucose significantly increased the pulsatile frequency and cellular volumes of cultured cardiomyocytes via PKC/NF-kappaB/c-fos pathway, which might lead to diabetic cardiomyopathy. Glucose 17-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 18854993-7 2009 In group 2 rats whose monocular (left eye) spiking was blocked by tetrodotoxin, a sodium channel blocker, prior to the unilateral (right eye) ON crush, the location of c-Fos expression was observed to be shifted to the side of LGN contralateral to the crush, while the expression pattern of c-Jun resembled the pattern of group 1 rats. Tetrodotoxin 66-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-173 19094973-0 2009 Behavioural disturbances and altered Fos protein expression in adult rats after chronic pubertal cannabinoid treatment. Cannabinoids 97-108 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 19094973-9 2009 Additionally, when comparing Fos IR in selected brain regions, these animals displayed altered basal neuronal activity and responded differently to acute application of haloperidol or apomorphine. Haloperidol 169-180 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 19094973-9 2009 Additionally, when comparing Fos IR in selected brain regions, these animals displayed altered basal neuronal activity and responded differently to acute application of haloperidol or apomorphine. Apomorphine 184-195 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 19200064-6 2009 In PvTh there was c-Fos IR in CTb-positive neurons associated with renewal showing activation of a PvTh-AcbSh pathway during renewal. pvth 3-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 19200064-6 2009 In PvTh there was c-Fos IR in CTb-positive neurons associated with renewal showing activation of a PvTh-AcbSh pathway during renewal. acbsh 104-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 19397204-0 2009 [Effects of curcumin on malondialdehyde and c-fos protein in hypoxia ischemia brain tissue in rats]. Curcumin 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 19397204-1 2009 OBJECTIVE: To explore the effects of curcumin on the content of malondialdehyde (MDA) and the expression level of c-fos protein following hypoxia ischemia brain damage (HIBD) in rats. Curcumin 37-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 19397204-7 2009 The expression level of c-fos protein was higher in the curcumin group than that in the other groups (P<0.05). Curcumin 56-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 19397204-9 2009 CONCLUSION: Curcumin could significantly decrease the content of MDA, increase the expression level of c-fos protein and reduce the damage of the neuron cells. Curcumin 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 18616988-6 2009 Microelectrode array recordings revealed that oxygen-glucose deprivation enhances hippocampal network firing rates, which induces c-fos transcription through a signaling pathway that, in contrast to Clca1, is activated by synaptic but not extrasynaptic NMDA receptors. oxygen-glucose 46-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-135 18955037-0 2009 Proximal colon distension induces Fos expression in oxytocin-, vasopressin-, CRF- and catecholamines-containing neurons in rat brain. Catecholamines 86-100 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 18762217-5 2009 Using c-fos immunoreactivity (FOS-IR) as a marker of neuronal activation in rats, we observed that mCPP significantly and dose-dependently activated a discrete population of caudal NTS neurons at the level of the area postrema (AP). 1-(3-chlorophenyl)piperazine 99-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 6-11 19536504-4 2009 For that, c-Fos immunopositive cells were counted along the brainstem in rats intravenously infused with the selective NHE3 inhibitor AVE1599. ave1599 134-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-15 19536505-0 2009 Nitric oxide in the solitary tract nucleus (STn) modulates glucose homeostasis and FOS-ir expression after carotid chemoreceptor stimulation. Nitric Oxide 0-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-86 19536505-7 2009 FOS-ir expression in brainstem cells suggests that CChrc stimulation activates nitroxidergic pathways in the STn to regulate peripheral and central glucose homeostasis. cchrc 51-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 19796495-11 2009 Also, in some SB623 grafted rats that were sacrificed within 2 h of dl-amphetamine injection, hot spots of c-Fos-positive nuclei that coincided with rejuvenated dense TH fibers around the grafted SB623 cells were observed, suggesting increased availability of DA in these locations. Amphetamine 68-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 107-112 21783926-0 2009 Cyhalothrin increased c-fos immunoreactivity at the paraventricular nucleus of the hypothalamus in rats, and suppressed macrophage activity in an adrenal-dependent fashion. cyhalothrin 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 21783926-3 2009 Results showed that cyhalothrin treatment (3.0mg/kg/day, for 7 days) increased corticosterone serum levels and c-fos immunoreactivity at the paraventricular nucleus of the hypothalamus (PVN) but induced no changes in c-fos expression at the basolateral amygdala (BLA). cyhalothrin 20-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 21783926-3 2009 Results showed that cyhalothrin treatment (3.0mg/kg/day, for 7 days) increased corticosterone serum levels and c-fos immunoreactivity at the paraventricular nucleus of the hypothalamus (PVN) but induced no changes in c-fos expression at the basolateral amygdala (BLA). cyhalothrin 20-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 217-222 18661553-6 2009 Moreover, we demonstrated that AP-1 (i.e., c-Fos/c-Jun) is crucial for oxLDL-induced proMMP-9 expression which was attenuated by pretreatment with AP-1 inhibitor (curcumin). Curcumin 163-171 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 43-48 19010489-5 2009 Spinal c-fos expression induced by 0.5% acetic acid was investigated after 0.1 mg/kg GRC-6211 or vehicle administration. Acetic Acid 40-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 7-12 19010489-11 2009 The c-fos expression induced by acetic acid was decreased by GRC-6211 (85.5 +/- 19.1 to 46.7 +/- 9.4, p <0.05). Acetic Acid 32-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 17943458-4 2009 Incubation of rat stomach tissue with NAA 1.5 mM, 1.5 microM and 1.5 nM induced inflammatory agents TNFalpha, p38MAPK, iNOS, PKC, COX2 and ICAM3; transcription factors phospho-NF-kBp65, cjun and cfos; contractile proteins MLCK and phospho MLC; and calcium channel alpha1C and calcium channel, voltage-dependent, beta 3 subunit compared to their respective control. N-acetylaspartate 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 195-199 18655797-3 2009 We found that Fos expression in LH orexin neurons varied in proportion to preference for morphine, cocaine or food. Morphine 89-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 18655797-3 2009 We found that Fos expression in LH orexin neurons varied in proportion to preference for morphine, cocaine or food. Cocaine 99-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 19898681-8 2009 After a noxious stimulus (e.g., capsaicin injection) or tissue injury, c-Fos begins to be induced after 30-60 minutes, whereas pERK can be induced within a minute, which can correlate well with the development of pain hypersensitivity. Capsaicin 32-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 18693129-0 2009 Effects of zonisamide on c-Fos expression under conditions of tacrine-induced tremulous jaw movements in rats: a potential mechanism underlying its anti-parkinsonian tremor effect. Zonisamide 11-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 18693129-0 2009 Effects of zonisamide on c-Fos expression under conditions of tacrine-induced tremulous jaw movements in rats: a potential mechanism underlying its anti-parkinsonian tremor effect. Tacrine 62-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 18693129-5 2009 Zonisamide suppressed the tacrine-induced c-Fos expression in the cortex, the dorsal striatum, and the nucleus accumbens, which are involved in the architecture of the cortico-basal ganglia-thalamocortical circuits. Zonisamide 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 18693129-5 2009 Zonisamide suppressed the tacrine-induced c-Fos expression in the cortex, the dorsal striatum, and the nucleus accumbens, which are involved in the architecture of the cortico-basal ganglia-thalamocortical circuits. Tacrine 26-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 18996447-0 2009 The distribution of gamma-hydroxybutyrate-induced Fos expression in rat brain: comparison with baclofen. Sodium Oxybate 20-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 18996447-5 2009 The present study used Fos immunohistochemistry to assess the neural activation produced in rat brain by medium to high doses of GHB (250, 500 and 1000 mg/kg) and a high dose of baclofen (10 mg/kg) that produced similar sedation to 500 mg/kg GHB. Sodium Oxybate 129-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 23-26 19003793-10 2009 Formalin testing evoked c-fos expression in these pontospinal neurons, suggesting that they were nociresponsive (A5-21%, LC-16%, and A7-26%, n = 8). Formaldehyde 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 19129396-7 2009 DMH NPY knockdown increased the feeding inhibitory and NTS c-Fos responses to peripheral administration of cholecystokinin. 1,2-Dimethylhydrazine 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 19121572-7 2009 Besides, for the nitroglycerin-induced headache rats, the c-fos gene expression in the brain stem and hypothalamus was remarkably inhibited and the level of plasma CGRP was reduced significantly after CXVO administration at both doses 90.0 and 135.0 microg/kg. Nitroglycerin 17-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 19194492-8 2009 Indeed, c-Fos induction by increasing concentrations of the GABAA antagonist picrotoxin in visual cortical slices was similar between PV cells and the other cortical neurons. Picrotoxin 77-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-13 18762914-7 2009 GSK207040 (3.2 mg/kg) raised extracellular concentrations of dopamine, noradrenaline, and acetylcholine in the anterior cingulate cortex and c-fos expression in the core of the nucleus accumbens was increased at doses of 3.2 and 10.0 mg/kg. 5-((3-cyclobutyl-2,3,4,5-tetrahydro-1H-3-benzazepin-7-yl)oxy)-N-methyl-2-pyrazinecarboxamide 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 19041662-0 2009 In vivo study on the effects of microcystin extracts on the expression profiles of proto-oncogenes (c-fos, c-jun and c-myc) in liver, kidney and testis of male Wistar rats injected i.v. microcystin 32-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 18805459-6 2008 The mean number of c-Fos positive neurons in the layers II-III began to increase at 1h and reached a peak at 2h after BV treatment that was followed by a gradual decrease afterward. Hydrogen 84-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-24 18561989-0 2009 Effect of long-term oxybutynin administration on c-Fos expression in spinal neurons: inhibition of antimuscarinics on bladder afferents in conscious rats. oxybutynin 20-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 18561989-1 2009 OBJECTIVES: To investigate the effect of long-term administration of oxybutynin on afferent input from the bladder by evaluating c-Fos expression in the spinal cord. oxybutynin 69-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 18561989-7 2009 In the rats that received low-dose oxybutynin, the number of c-Fos-positive neurons in the spinal cord was significantly lower than that in controls. oxybutynin 35-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 18561989-8 2009 CONCLUSIONS: Administration of low-dose oxybutynin decreased the c-Fos expression induced by continuous infusion of saline into the bladder. oxybutynin 40-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 18561989-8 2009 CONCLUSIONS: Administration of low-dose oxybutynin decreased the c-Fos expression induced by continuous infusion of saline into the bladder. Sodium Chloride 116-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 18805459-4 2008 In naive and saline-treated rats, c-Fos-labeled neurons were diffusely and sparsely distributed in the hindlimb region of S1 area. Sodium Chloride 13-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 18950690-8 2008 Nicotine increased Fos expression in orexin neurons projecting to both basal forebrain and PVT. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 19077321-11 2008 The number of Fos-ir neurons in the OVX-CT-pcpa-group was significantly more than that of the OVX-CT-group (p = 0.0283), which means pcpa inhibits calcitonin induced reduction of c-Fos production. ovx-ct-pcpa 36-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 19077321-11 2008 The number of Fos-ir neurons in the OVX-CT-pcpa-group was significantly more than that of the OVX-CT-group (p = 0.0283), which means pcpa inhibits calcitonin induced reduction of c-Fos production. ovx-ct-pcpa 36-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-184 19077321-11 2008 The number of Fos-ir neurons in the OVX-CT-pcpa-group was significantly more than that of the OVX-CT-group (p = 0.0283), which means pcpa inhibits calcitonin induced reduction of c-Fos production. Fenclonine 43-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 18822274-0 2008 Effects of dopamine and NMDA receptors on cocaine-induced Fos expression in the striatum of Fischer rats. Dopamine 11-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 18938224-7 2008 2-[2-(2-Methoxy-1,4-benzodioxanyl)]imidazoline hydrochloride (RX821002), an alpha-2 adrenergic receptor (A2AR) antagonist, administered to control PND 60 animals produces elevations of Arc, zif268 and c-fos mRNAs. RX 821002 hydrochloride 0-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 201-206 19107124-8 2009 Levels of Fos protein in a number of spinally projecting neurons within discrete hypothalamic and brainstem sites were elevated in olanzapine-treated rats. Olanzapine 131-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-13 18822274-0 2008 Effects of dopamine and NMDA receptors on cocaine-induced Fos expression in the striatum of Fischer rats. Cocaine 42-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 18822274-2 2008 In this study, we show that a single cocaine administration (30 mg/kg) time-dependently increases ERK phosphorylation, c-Fos and FosB protein expression, and MKP-1 phosphorylation (p-MKP-1), in the caudate-putamen (CPu) and nucleus accumbens (NAc) of Fischer rats. Cocaine 37-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 18822274-3 2008 In the CPu, 1 h after cocaine injection, the increase in c-Fos and FosB protein expressions is totally abolished by pre-administration of DA-D1 receptor antagonist, SCH23390. Cocaine 22-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 18822274-3 2008 In the CPu, 1 h after cocaine injection, the increase in c-Fos and FosB protein expressions is totally abolished by pre-administration of DA-D1 receptor antagonist, SCH23390. SCH 23390 165-173 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 18822274-4 2008 In the NAc, SCH23390 also inhibits cocaine-induced c-Fos protein expression. SCH 23390 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 18822274-4 2008 In the NAc, SCH23390 also inhibits cocaine-induced c-Fos protein expression. Cocaine 35-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 18822274-5 2008 The pre-treatment of NMDA receptor antagonist, MK801, partially reduces cocaine-activated c-Fos protein expression in the CPu. Dizocilpine Maleate 47-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 18840509-4 2008 We examined the CQ induced loss of synaptic zinc, cell death and c-Fos induction in rats and mice. Clioquinol 16-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 19120104-11 2008 In saline-treated rats, maximum activation of Fos reached around 4% in +/+, 20% in di/+ rats, and as much as 60% in di/di rats. Sodium Chloride 3-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 19120105-2 2008 Kainic acid or egg white treatment weakly activated Fos expression in the PVH of lactating in comparison to diestrus or ovariectomized (OVX) rats. Kainic Acid 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 19120132-5 2008 We show here that positive modulators of alpha(1)-subtype containing GABA-A receptors with zolpidem (10 mg/kg) increase HPA activity in terms of increase in plasma corticosterone and induction of Fos in the PVN, whereas activation of non-alpha(1)-subtype GABA-A receptors using L-818,417 (10 mg/kg) likely inhibits the activity. Zolpidem 91-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 196-199 18846040-3 2008 dexmedetomidine and ST-91 in the formalin behavioural model and their effects on primary afferent substance P (SP) release and spinal Fos activation. Dexmedetomidine 0-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-137 18846040-3 2008 dexmedetomidine and ST-91 in the formalin behavioural model and their effects on primary afferent substance P (SP) release and spinal Fos activation. ST 91 20-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-137 18846040-9 2008 Finally, the effects of dexmedetomidine on formalin-induced Fos expression were assessed in the dorsal horn. Formaldehyde 43-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-63 18846040-13 2008 On the other hand, both dexmedetomidine and morphine diminished the formalin-induced Fos activation. Dexmedetomidine 24-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 18846040-13 2008 On the other hand, both dexmedetomidine and morphine diminished the formalin-induced Fos activation. Morphine 44-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 18846040-13 2008 On the other hand, both dexmedetomidine and morphine diminished the formalin-induced Fos activation. Formaldehyde 68-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 18846040-14 2008 The effect of dexmedetomidine on formalin-induced Fos activation was reversed by BRL44408, but not ARC239. Dexmedetomidine 14-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 18846040-14 2008 The effect of dexmedetomidine on formalin-induced Fos activation was reversed by BRL44408, but not ARC239. Formaldehyde 33-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 18787053-12 2008 These results indicate that oxidative stress plays an important role in cardiac hypertrophy, and suggest redox activation of AKT1 and JUN/FOS signaling pathways with H2O2 acting as a possible intracellular mediator in this adaptive response to experimental hyperthyroidism. Hydrogen Peroxide 166-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-141 19000373-9 2008 Furthermore, isorhynchophylline increased the NO content and NOS activity, and suppressed Ang II-induced over-expression of c-fos, OPN and PCNA. rhyncophylline 13-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-129 19000373-10 2008 Thus, isorhynchophylline was effective against Ang-II induced cell proliferation, an effect that appears to be due, at least in part, to increased NO production, regulation of the cell cycle, and depressed expression of c-fos, OPN and PCNA related to VMSC proliferation. rhyncophylline 6-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 220-225 18792992-0 2008 Differential expression of Fos and Zif268 in the nigrostriatal system after methamphetamine administration in a rat model of Parkinson"s disease. Methamphetamine 76-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 18792992-1 2008 The goal of this study was to examine the topological specificity of methamphetamine-induced activation of the immediate-early gene proteins, Fos and Zif268, in the nigrostriatal system in a unilateral 6-hydroxydopamine (6-OHDA) rat model of Parkinson"s disease with or without intrastriatal grafts of fetal ventral mesencephalon. Methamphetamine 69-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-145 18792992-1 2008 The goal of this study was to examine the topological specificity of methamphetamine-induced activation of the immediate-early gene proteins, Fos and Zif268, in the nigrostriatal system in a unilateral 6-hydroxydopamine (6-OHDA) rat model of Parkinson"s disease with or without intrastriatal grafts of fetal ventral mesencephalon. Oxidopamine 202-219 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-145 18792992-1 2008 The goal of this study was to examine the topological specificity of methamphetamine-induced activation of the immediate-early gene proteins, Fos and Zif268, in the nigrostriatal system in a unilateral 6-hydroxydopamine (6-OHDA) rat model of Parkinson"s disease with or without intrastriatal grafts of fetal ventral mesencephalon. Oxidopamine 221-227 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-145 18792992-3 2008 induced Fos-like immunoreactivity (FLI) dominantly in the striatum and the globus pallidus (GP) on the intact side as well as in the substantia nigra pars reticulata (SNr) on the lesioned side in the 6-OHDA rats. Oxidopamine 200-206 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-11 18792992-5 2008 In the striatum, a similar tendency could be observed between Fos and Zif268 immunoreactivity following methamphetamine. Methamphetamine 104-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 62-65 18792992-8 2008 The differential expression of Fos and Zif268 was observed among the three regions of the nigrostriatal system following methamphetamine in the 6-OHDA rats. Methamphetamine 121-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 18792992-8 2008 The differential expression of Fos and Zif268 was observed among the three regions of the nigrostriatal system following methamphetamine in the 6-OHDA rats. Oxidopamine 144-150 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 19180910-6 2008 c-Fos/ NADPH-d expression was observed in the ipsilesional MVN and the contralesional PrH of UVD rats. NADP 7-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 19180910-8 2008 CONCLUSION: In the ipsilesional MVN and the contralesional PrH, c-Fos plays an important role in vestibular compensation, in which nitric oxide acts as a key neurotransmitter. Nitric Oxide 131-143 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 19288899-9 2008 The number of c-fos positive neurons in the I-II layers of cSTN was obviously increased following EA at ST2, being significant more than that in control, vehicle, capsaicin and sham groups (P<0.01 respectively). Capsaicin 163-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 19288899-11 2008 In capsaicin + EA group, the number of c-fos-IR positive neurons in the I-II layers of cSTN was obviously less than that in vehicle and vehicle + EA groups (P<0.01), but no significant changes were found in III-IV layers of cSTN. Capsaicin 3-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 18930597-0 2008 Serotonin 5-HT2A receptor involvement and Fos expression at the spinal level in vincristine-induced neuropathy in the rat. Vincristine 80-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 18930597-8 2008 Immunocytochemical study of Fos expression in vincristine-treated rats revealed a significant increase in the number of Fos-positive neurons not only in regions where nociceptive fibres terminate superficial (I-II) and deep layers (V-VI) of the spinal cord, but also in intermediate layers, suggesting that Abeta fibres could be involved in the spinal sensitization observed in this model. Vincristine 46-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 18930597-8 2008 Immunocytochemical study of Fos expression in vincristine-treated rats revealed a significant increase in the number of Fos-positive neurons not only in regions where nociceptive fibres terminate superficial (I-II) and deep layers (V-VI) of the spinal cord, but also in intermediate layers, suggesting that Abeta fibres could be involved in the spinal sensitization observed in this model. Vincristine 46-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-123 18930597-9 2008 Double labelling experiments showed that Fos-positive neurons were endowed with 5-HT2AR immunolabelling in the dorsal horn of vincristine-treated rats. Vincristine 126-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 18789920-0 2008 Delta-9-tetrahydrocannabinol differently affects striatal c-Fos expression following haloperidol or clozapine administration. Dronabinol 0-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 18789920-0 2008 Delta-9-tetrahydrocannabinol differently affects striatal c-Fos expression following haloperidol or clozapine administration. Haloperidol 85-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 18789920-0 2008 Delta-9-tetrahydrocannabinol differently affects striatal c-Fos expression following haloperidol or clozapine administration. Clozapine 100-109 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 18789920-3 2008 Western Blot and immunocytochemistry stereological analyses indicated that delta-9-tetrahydrocannabinol (0.5 mg/kg) increased striatal c-Fos immunoreactivity induced by haloperidol (0.1 mg/kg). Dronabinol 75-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 18789920-3 2008 Western Blot and immunocytochemistry stereological analyses indicated that delta-9-tetrahydrocannabinol (0.5 mg/kg) increased striatal c-Fos immunoreactivity induced by haloperidol (0.1 mg/kg). Haloperidol 169-180 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-140 18789920-5 2008 The present results indicate that the behavioral effects induced by delta-9-tetrahydrocannabinol in haloperidol- and clozapine-treated rats are associated with different striatal c-Fos expressions. Dronabinol 68-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-184 18789920-5 2008 The present results indicate that the behavioral effects induced by delta-9-tetrahydrocannabinol in haloperidol- and clozapine-treated rats are associated with different striatal c-Fos expressions. Haloperidol 100-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-184 18789920-5 2008 The present results indicate that the behavioral effects induced by delta-9-tetrahydrocannabinol in haloperidol- and clozapine-treated rats are associated with different striatal c-Fos expressions. Clozapine 117-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 179-184 18838113-8 2008 Four experiments with unilateral clonidine injections into the A7 region and with Fos immunohistochemistry used as a marker of cell activity revealed that the percentage of Fos-positive A7 cells was significantly reduced on the injected side. Clonidine 33-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 173-176 18848603-0 2008 Acute nicotine activates c-fos and activity-regulated cytoskeletal associated protein mRNA expression in limbic brain areas involved in the central stress-response in rat pups during a period of hypo-responsiveness to stress. Nicotine 6-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 18848603-9 2008 Acute nicotine resulted in significant induction of c-fos expression in the PVN and CeA at P5, P7 and P10, and in the BST at P7 and P10. Nicotine 6-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 18822353-0 2008 The anandamide membrane transporter inhibitor, VDM-11, modulates sleep and c-Fos expression in the rat brain. N-(2-methyl-3-hydroxyphenyl)-5,8,11,14-eicosatetraenamide 47-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 18822353-9 2008 Additionally, VDM-11 enhanced c-Fos expression in sleep-related brain areas such as the anterior hypothalamic area, paraventricular thalamic nucleus, and pedunculopontine tegmental nucleus. N-(2-methyl-3-hydroxyphenyl)-5,8,11,14-eicosatetraenamide 14-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 18784083-1 2008 It has been demonstrated that c-Fos has, in addition to its well recognized AP-1 transcription factor activity, the capacity to associate to the endoplasmic reticulum and activate key enzymes involved in the synthesis of phospholipids required for membrane biogenesis during cell growth and neurite formation. Phospholipids 221-234 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 18784083-2 2008 Because membrane genesis requires the coordinated supply of all its integral membrane components, the question emerges as to whether c-Fos also activates the synthesis of glycolipids, another ubiquitous membrane component. Glycolipids 171-182 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-138 18784083-4 2008 Specifically, c-Fos activates the enzyme glucosylceramide synthase (GlcCerS), the product of which, GlcCer, is the first glycosylated intermediate in the pathway of synthesis of glycolipids. Glycolipids 178-189 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 18784337-8 2008 The distension increased the number of Fos- and double-labeled Fos/CRF neurons in the parvocellular PVN, which was higher in the water-deprived than in the hydrated group (P < 0.01). Water 129-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-42 18784337-8 2008 The distension increased the number of Fos- and double-labeled Fos/CRF neurons in the parvocellular PVN, which was higher in the water-deprived than in the hydrated group (P < 0.01). Water 129-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 18973603-0 2008 Acute and sensitized response to 3,4-methylenedioxymethamphetamine in rats: different behavioral profiles reflected in different patterns of Fos expression. N-Methyl-3,4-methylenedioxyamphetamine 33-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-144 18755745-8 2008 ICA infusions of hypertonic NaCl and mannitol each significantly (P < 0.01-0.001) increased the number of Fos immunoreactive (Fos-ir) neuronal nuclei in the dorsal cap (DC) and lateral margins (LM) of OVLT. Mannitol 37-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-112 18755745-8 2008 ICA infusions of hypertonic NaCl and mannitol each significantly (P < 0.01-0.001) increased the number of Fos immunoreactive (Fos-ir) neuronal nuclei in the dorsal cap (DC) and lateral margins (LM) of OVLT. Mannitol 37-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-132 18755745-9 2008 In the LM, infusions of 1.5 and 3.0 osmol kg(-1) NaCl produced similar increases in the number of Fos-ir neurones. Sodium Chloride 49-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-101 18755745-11 2008 Among OVLT neurones with axons projecting directly to the PVN (i.e. CTB-ir), graded hypertonic NaCl infusions again produced graded increases in Fos expression and this was observed in both the DC and LM. Sodium Chloride 95-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-148 18755745-12 2008 Although the DC and LM contained a similar number of OVLT-PVN neurones, the proportion of such neurones that expressed Fos-ir in responses to ICA hypertonic NaCl infusions was greater in the DC (P < 0.001). Sodium Chloride 157-161 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-122 18656497-7 2008 GnRH activation, as measured by the percentage of immunopositive GnRH neurons that were also immunopositive for Fos, was significantly lower in all treatment groups except the DPN group compared to the control group. diarylpropionitrile 176-179 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 19102919-6 2008 CONCLUSION: High, especially intermittent high glucose could lead to diabetic cardiomyopathy by promoting cardiac hypertrophy, increasing beating frequency via activating PKC/NF-kappaB/c-fos pathways. Glucose 47-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 185-190 18755245-3 2008 In rats, we determined that yohimbine elicits patterns of brain expression of the mRNAs for c-fos, a marker of neuronal activation, and corticotropin-releasing factor (CRF) a stress-related peptide, distinct from that produced by FG-7142. Yohimbine 28-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-97 18755245-6 2008 As we found in the rat, yohimbine selectively increased c-fos mRNA in the mouse NACs, BLA and CeA. Yohimbine 24-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 56-61 18761334-3 2008 Delayed Fos expression in response to nitroglycerin (NTG) administration is a procedure used to identify the neuroanatomical substrates of the migraine condition. Nitroglycerin 38-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-11 18761334-3 2008 Delayed Fos expression in response to nitroglycerin (NTG) administration is a procedure used to identify the neuroanatomical substrates of the migraine condition. Nitroglycerin 53-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-11 18773942-9 2008 DSAP lesions depleted NA terminals in the PVN and bed nucleus of the stria terminalis, reduced the number of NA cell bodies in the NST and VLM, attenuated PVN Fos activation after LPS, and attenuated LPS-induced increases in plasma corticosterone. dsap 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 159-162 18988310-6 2008 Celecoxib prevented microglia activation in the hilus and inhibited the abnormal neurogenesis and astrogliosis in the hippocampus by inhibiting MAPK/ERK activity and c-Fos transcription. Celecoxib 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-171 19211972-0 2008 Inhibition of neophobia-stimulated c-Fos expression in the dorsomedial part of the prefrontal cortex in rats pretreated with midazolam. Midazolam 125-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 18973603-8 2008 Acute MDMA exposure produced a dose-dependent increase in locomotion in the peripheral zone of the open field that was related to an increase in Fos expression in the ventromedial shell of the nucleus accumbens, ventral pallidum, several hypothalamic nuclei and rhomboid thalamic nucleus. N-Methyl-3,4-methylenedioxyamphetamine 6-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-148 18765271-0 2008 Estradiol replacement modifies c-fos expression at the spinomedullary junction evoked by temporomandibular joint stimulation in ovariectomized female rats. Estradiol 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 18706486-10 2008 The abolition of Fos protein expression evoked by hypoxia suggested that caffeine exposure may decrease the activity of O2-sensing peripheral chemoreceptor pathway. Caffeine 73-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 18706486-10 2008 The abolition of Fos protein expression evoked by hypoxia suggested that caffeine exposure may decrease the activity of O2-sensing peripheral chemoreceptor pathway. Oxygen 120-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-20 18765271-4 2008 E2 treatment also modified the influence of N-methyl-D-aspartate (NMDA) and AMPA receptor antagonists on TMJ-evoked Fos-LI. N-Methylaspartate 44-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 18765271-4 2008 E2 treatment also modified the influence of N-methyl-D-aspartate (NMDA) and AMPA receptor antagonists on TMJ-evoked Fos-LI. N-Methylaspartate 66-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 18765271-5 2008 The NMDA antagonist, MK-801, dose-dependently reduced the Fos-LI response at the Vc/C1-2 junction in HE2 rats, while only high dose MK-801 was effective in LE2 rats. N-Methylaspartate 4-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 18765271-5 2008 The NMDA antagonist, MK-801, dose-dependently reduced the Fos-LI response at the Vc/C1-2 junction in HE2 rats, while only high dose MK-801 was effective in LE2 rats. Dizocilpine Maleate 21-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 18765271-6 2008 MK801 reduced equally the Fos-LI response at the Vi/Vc transition in both groups, while only minor effects were seen at the dPa5 region. Dizocilpine Maleate 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-29 18765271-7 2008 The AMPA receptor antagonist, NBQX, reduced Fos-LI at the Vc/C(1-2) and Vi/Vc(vl) regions in HE2 rats, while only high dose NBQX was effective in LE2 rats. 2,3-dioxo-6-nitro-7-sulfamoylbenzo(f)quinoxaline 30-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 18722360-0 2008 Inactivation of median preoptic nucleus causes c-Fos expression in hypocretin- and serotonin-containing neurons in anesthetized rat. Serotonin 83-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 18687381-0 2008 Effects of treadmill exercise on memory and c-Fos expression in the hippocampus of the rats with intracerebroventricular injection of streptozotocin. Streptozocin 134-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 18687381-7 2008 The results of the present study showed that ICV injection of STZ impaired long-term memory capacity and decreased the number of c-Fos-positive cells in several regions of the rat hippocampus. Streptozocin 62-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 18687381-8 2008 However, treadmill exercise alleviated long-term memory deficits and enhanced c-Fos expression in the rats with ICV injection of STZ. Streptozocin 129-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 18834549-0 2008 Amphetamine and pseudoephedrine cross-tolerance measured by c-Fos protein expression in brains of chronically treated rats. Pseudoephedrine 16-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 60-65 18834549-5 2008 (i) Chronic exposure to pseudoephedrine reduced the c-Fos response to acute pseudoephedrine treatment suggesting that pseudoephedrine induced tolerance in the animals. Pseudoephedrine 24-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 18834549-5 2008 (i) Chronic exposure to pseudoephedrine reduced the c-Fos response to acute pseudoephedrine treatment suggesting that pseudoephedrine induced tolerance in the animals. Pseudoephedrine 76-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 18834549-5 2008 (i) Chronic exposure to pseudoephedrine reduced the c-Fos response to acute pseudoephedrine treatment suggesting that pseudoephedrine induced tolerance in the animals. Pseudoephedrine 76-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 18834549-6 2008 (ii) In animals chronically treated with amphetamine or pseudoephedrine the acute c-Fos response to pseudoephedrine and amphetamine was reduced respectively as compared to naive animals indicating cross-tolerance for the two drugs. Amphetamine 41-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 18834549-6 2008 (ii) In animals chronically treated with amphetamine or pseudoephedrine the acute c-Fos response to pseudoephedrine and amphetamine was reduced respectively as compared to naive animals indicating cross-tolerance for the two drugs. Pseudoephedrine 56-71 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 18834549-6 2008 (ii) In animals chronically treated with amphetamine or pseudoephedrine the acute c-Fos response to pseudoephedrine and amphetamine was reduced respectively as compared to naive animals indicating cross-tolerance for the two drugs. Pseudoephedrine 100-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 18834549-6 2008 (ii) In animals chronically treated with amphetamine or pseudoephedrine the acute c-Fos response to pseudoephedrine and amphetamine was reduced respectively as compared to naive animals indicating cross-tolerance for the two drugs. Amphetamine 120-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 18722360-5 2008 We examined the effects of inactivation of MnPN neurons by locally microinjecting 0.2 microl of 1 mM or 10 mM solutions of a GABA(A) receptor agonist, muscimol, into the MnPN on Fos expression (Fos-IR) in the PF-LHA neurons including HCRT neurons and 5-HT neurons in the DRN in anesthetized rats. Muscimol 151-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 178-181 18722360-6 2008 Compared to artificial cerebrospinal fluid control, microinjection of muscimol into the MnPN resulted in significantly higher percentages of HCRT and non-HCRT neurons in the PF-LHA and 5-HT neurons in the DRN that exhibited Fos-IR. Muscimol 70-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 224-227 18650316-2 2008 Hydralazine (HDZ)-induced hypotension activates NK3R expressed by magnocellular neurons, increases plasma VP and OT levels, and induces c-Fos expression in VP and OT neurons. Hydralazine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 18667712-5 2008 To test this hypothesis, we mapped and quantified the Fos-immunoreactive response to 2DG in control and dehydrated rats drinking 2.5% saline. Deoxyglucose 85-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 18667712-5 2008 To test this hypothesis, we mapped and quantified the Fos-immunoreactive response to 2DG in control and dehydrated rats drinking 2.5% saline. Sodium Chloride 134-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 18667712-6 2008 Our rationale was that regions showing an attenuated Fos response to 2DG in dehydrated animals would be strong candidates as the targets of dehydration-induced suppression of 2DG feeding. Deoxyglucose 69-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 18650316-2 2008 Hydralazine (HDZ)-induced hypotension activates NK3R expressed by magnocellular neurons, increases plasma VP and OT levels, and induces c-Fos expression in VP and OT neurons. Hydralazine 13-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 136-141 18667712-6 2008 Our rationale was that regions showing an attenuated Fos response to 2DG in dehydrated animals would be strong candidates as the targets of dehydration-induced suppression of 2DG feeding. Deoxyglucose 175-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 18667712-7 2008 We found that the Fos response to combined dehydration and 2DG was attenuated only in the lateral hypothalamic area, with dehydration alone increasing Fos in the lateral part of the paraventricular nucleus. Deoxyglucose 59-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 18650316-10 2008 Intra-PVN injection of SB-222200 prior to an intravenous injection of HDZ significantly decreased c-Fos expression in both VP and OT neurons by approximately 70% and attenuated plasma VP and OT levels by 33% and 35%, respectively. SB 222200 23-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 98-103 18667712-7 2008 We found that the Fos response to combined dehydration and 2DG was attenuated only in the lateral hypothalamic area, with dehydration alone increasing Fos in the lateral part of the paraventricular nucleus. Deoxyglucose 59-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-154 18667712-8 2008 In the arcuate nucleus and those regions of the hindbrain that provide afferent inputs critical for the feeding response to 2DG, the Fos response to 2DG was unaffected by dehydration. Deoxyglucose 149-152 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 133-136 18685071-4 2008 High testosterone replacement decreased plasma adrenocorticotropin hormone (ACTH) and paraventricular nucleus (PVN) Fos responses to restraint exposure in sham- but not in MPN-lesioned animals. Testosterone 5-17 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 116-119 18753261-0 2008 Estradiol replacement enhances sleep deprivation-induced c-Fos immunoreactivity in forebrain arousal regions of ovariectomized rats. Estradiol 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 18753261-1 2008 To understand how female sex hormones influence homeostatic mechanisms of sleep, we studied the effects of estradiol (E(2)) replacement on c-Fos immunoreactivity in sleep/wake-regulatory brain areas after sleep deprivation (SD) in ovariectomized rats. Estradiol 118-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 139-144 18823165-0 2008 A morphine-paired environment alters c-Fos expression in the forebrain of rats displaying conditioned place preference or aversion. Morphine 2-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 18823165-6 2008 Compared with the control group, the CPP and CPA groups showed a significant increase of c-Fos expression in the dorsomedial striatum, central medial nucleus of the thalamus, and the basolateral amygdala. cpa 45-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 18637015-3 2008 Immunocytochemistry and histochemistry were performed to determine the effects of emulsified isoflurane on formalin-induced changes in Fos-like immunoreactive (Fos-LI)- and nicotinamide adenine dinucleotide phosphatediaphorase (NADPH-d)-positive neurons, respectively. Isoflurane 93-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-138 18637015-3 2008 Immunocytochemistry and histochemistry were performed to determine the effects of emulsified isoflurane on formalin-induced changes in Fos-like immunoreactive (Fos-LI)- and nicotinamide adenine dinucleotide phosphatediaphorase (NADPH-d)-positive neurons, respectively. Isoflurane 93-103 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-163 18637015-3 2008 Immunocytochemistry and histochemistry were performed to determine the effects of emulsified isoflurane on formalin-induced changes in Fos-like immunoreactive (Fos-LI)- and nicotinamide adenine dinucleotide phosphatediaphorase (NADPH-d)-positive neurons, respectively. Formaldehyde 107-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 135-138 18637015-3 2008 Immunocytochemistry and histochemistry were performed to determine the effects of emulsified isoflurane on formalin-induced changes in Fos-like immunoreactive (Fos-LI)- and nicotinamide adenine dinucleotide phosphatediaphorase (NADPH-d)-positive neurons, respectively. Formaldehyde 107-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-163 18637015-5 2008 Furthermore, Fos-LI- and NADPH-d-positive neurons were mainly found in the ipsilateral dorsal horn after injection of formalin, some of which were Fos-LI/NADPH-d double-labelled neurons. NADP 25-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-150 18637015-5 2008 Furthermore, Fos-LI- and NADPH-d-positive neurons were mainly found in the ipsilateral dorsal horn after injection of formalin, some of which were Fos-LI/NADPH-d double-labelled neurons. Formaldehyde 118-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 18637015-5 2008 Furthermore, Fos-LI- and NADPH-d-positive neurons were mainly found in the ipsilateral dorsal horn after injection of formalin, some of which were Fos-LI/NADPH-d double-labelled neurons. Formaldehyde 118-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-150 18637015-5 2008 Furthermore, Fos-LI- and NADPH-d-positive neurons were mainly found in the ipsilateral dorsal horn after injection of formalin, some of which were Fos-LI/NADPH-d double-labelled neurons. [[(2R,3R,4R,5R)-5-(6-aminopurin-9-yl)-3-hydroxy-4-phosphonooxyoxolan-2-yl]methoxy-hydroxyphosphoryl] [(2R,3S,4R,5R)-5-(3-carbamoyl-4-deuterio-4H-pyridin-1-yl)-3,4-dihydroxyoxolan-2-yl]methyl hydrogen phosphate 25-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-150 18637015-6 2008 Administration of emulsified isofluane significantly decreased Fos-LI- and NADPH-d-positive, as well as Fos-LI/NADPH-d double-labelled, neurons. isofluane 29-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 18637015-6 2008 Administration of emulsified isofluane significantly decreased Fos-LI- and NADPH-d-positive, as well as Fos-LI/NADPH-d double-labelled, neurons. isofluane 29-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 18637015-6 2008 Administration of emulsified isofluane significantly decreased Fos-LI- and NADPH-d-positive, as well as Fos-LI/NADPH-d double-labelled, neurons. Deuterium 1-2 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 18637015-6 2008 Administration of emulsified isofluane significantly decreased Fos-LI- and NADPH-d-positive, as well as Fos-LI/NADPH-d double-labelled, neurons. Deuterium 1-2 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 18637015-6 2008 Administration of emulsified isofluane significantly decreased Fos-LI- and NADPH-d-positive, as well as Fos-LI/NADPH-d double-labelled, neurons. Deuterium 27-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-66 18637015-6 2008 Administration of emulsified isofluane significantly decreased Fos-LI- and NADPH-d-positive, as well as Fos-LI/NADPH-d double-labelled, neurons. Deuterium 27-28 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 104-107 18812028-7 2008 Moreover, UA significantly suppressed the arthritis-induced mechanical and thermal hyperalgesia as well as the spinal Fos expression, as determined by immunohistochemistry, which was increased by CFA injection. ursolic acid 10-12 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 18548233-4 2008 We show that naloxone-induced morphine withdrawal activates extracellular signal-regulated kinases(1/2) and increases c-Fos expression in rat paraventricular nucleus and nucleus tractus solitarius-A(2) neurons. Naloxone 13-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 18548233-4 2008 We show that naloxone-induced morphine withdrawal activates extracellular signal-regulated kinases(1/2) and increases c-Fos expression in rat paraventricular nucleus and nucleus tractus solitarius-A(2) neurons. Morphine 30-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 18619501-3 2008 RTX treatment significantly attenuated BmK venom-induced c-Fos expression in all laminaes of bilateral L4-L5 lumbar spinal cord, especially in superficial laminaes. resiniferatoxin 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 18094667-7 2008 Forebrain neurons exhibiting both Fos and tracer immunoreactivity were enriched in both cocaine groups relative to the controls only in the globus pallidus and ventral pallidum, which, together, represented a minor part of total forebrain retrogradely labeled neurons. Cocaine 88-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-37 18185499-8 2008 The effects of SalvA on locomotor activity paralleled its effects on cocaine-induced c-Fos expression in the dorsal striatum: acute SalvA attenuated cocaine-induced c-Fos, whereas repeated SalvA potentiated it when administered in the activity chambers but not the home cage. Cocaine 69-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 18185499-8 2008 The effects of SalvA on locomotor activity paralleled its effects on cocaine-induced c-Fos expression in the dorsal striatum: acute SalvA attenuated cocaine-induced c-Fos, whereas repeated SalvA potentiated it when administered in the activity chambers but not the home cage. Cocaine 69-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 18185499-8 2008 The effects of SalvA on locomotor activity paralleled its effects on cocaine-induced c-Fos expression in the dorsal striatum: acute SalvA attenuated cocaine-induced c-Fos, whereas repeated SalvA potentiated it when administered in the activity chambers but not the home cage. Cocaine 149-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 165-170 18692030-0 2008 Ethanol-induced alterations of c-Fos immunoreactivity in specific limbic brain regions following ethanol discrimination training. Ethanol 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 18692030-0 2008 Ethanol-induced alterations of c-Fos immunoreactivity in specific limbic brain regions following ethanol discrimination training. Ethanol 97-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 18692030-3 2008 To accomplish this goal, immunohistochemistry was used to assess the effects of ethanol (2 g/kg) on c-Fos immunoreactivity (Fos-IR). Ethanol 80-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 18793388-6 2008 Serotonergic depletion of the lumbar spinal cord with 5,7 di-hydroxytryptamine creatinine sulphate (5,7-DHT) reduced the inflammation evoked P-MeCP2 in the superficial dorsal horn by 57%, and that of Zif268 and Fos by 37.5% and 30% respectively. 5,7 di-hydroxytryptamine creatinine sulphate 54-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 211-214 18793388-6 2008 Serotonergic depletion of the lumbar spinal cord with 5,7 di-hydroxytryptamine creatinine sulphate (5,7-DHT) reduced the inflammation evoked P-MeCP2 in the superficial dorsal horn by 57%, and that of Zif268 and Fos by 37.5% and 30% respectively. 5,7-dihydroxytryptamine creatinine 100-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 211-214 18639534-2 2008 Expression of c-Fos protein and pERK is mediated by the excitatory neurotransmitter, glutamate. Glutamic Acid 85-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 18606162-7 2008 The levels of transcription factors IRF-1 and c-Fos, as well as the phosphorylation of c-Jun were also reduced in adenosine-treated C6 cells, while the activation of NF-kappaB was enhanced via increased phosphorylation of its inhibitory unit IkappaB. Adenosine 114-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-51 18635449-0 2008 Expression of Fos during sham sucrose intake in rats with central gustatory lesions. Sucrose 30-37 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 18635449-8 2008 In controls, compared with water, sham ingesting sucrose produced significantly more Fos-positive neurons in the nucleus of the solitary tract, PBN, TTA, and gustatory cortex (GC). Sucrose 49-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 18635449-9 2008 In the ventral forebrain, sucrose sham licking increased Fos in the bed nucleus of the stria terminalis, central nucleus of amygdala, and the shell of nucleus accumbens. Sucrose 26-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 18635449-11 2008 After lateral PBN lesions, the Fos distributions produced by distilled H(2)O or sucrose intake did not differ from controls. Water 71-76 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 18635449-11 2008 After lateral PBN lesions, the Fos distributions produced by distilled H(2)O or sucrose intake did not differ from controls. Sucrose 80-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 18635449-12 2008 Bilateral medial PBN damage, however, eliminated the sucrose-induced Fos increase not only in the TTA and GC but also in the ventral forebrain. Sucrose 53-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 18488248-4 2008 Using an animal model of relapse to cocaine-seeking, the present study investigated the expression patterns of three different activity-related genes (c-fos, zif/268, and arc) in cortical and striatal brain regions implicated in compulsive drug-seeking in order to determine the neuroadaptations that occur during context-induced relapse following brief or prolonged abstinence from cocaine self-administration. Cocaine 383-390 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-156 18663473-3 2008 Fos-ir was assessed after intra-pallidal injections of the excitatory amino acid agonist, NMDA, or the GABA(A) antagonist, bicuculline in normal rats and in those rendered Parkinsonian-like by lesioning dopaminergic neurons with the neurotoxin, 6-OHDA. Excitatory Amino Acids 59-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 18663473-3 2008 Fos-ir was assessed after intra-pallidal injections of the excitatory amino acid agonist, NMDA, or the GABA(A) antagonist, bicuculline in normal rats and in those rendered Parkinsonian-like by lesioning dopaminergic neurons with the neurotoxin, 6-OHDA. N-Methylaspartate 90-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 18663473-3 2008 Fos-ir was assessed after intra-pallidal injections of the excitatory amino acid agonist, NMDA, or the GABA(A) antagonist, bicuculline in normal rats and in those rendered Parkinsonian-like by lesioning dopaminergic neurons with the neurotoxin, 6-OHDA. gamma-Aminobutyric Acid 103-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 18663473-3 2008 Fos-ir was assessed after intra-pallidal injections of the excitatory amino acid agonist, NMDA, or the GABA(A) antagonist, bicuculline in normal rats and in those rendered Parkinsonian-like by lesioning dopaminergic neurons with the neurotoxin, 6-OHDA. Bicuculline 123-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 18663473-3 2008 Fos-ir was assessed after intra-pallidal injections of the excitatory amino acid agonist, NMDA, or the GABA(A) antagonist, bicuculline in normal rats and in those rendered Parkinsonian-like by lesioning dopaminergic neurons with the neurotoxin, 6-OHDA. Oxidopamine 245-251 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 18663473-5 2008 Blocking tonically activated GABA(A) receptors with bicuculline (50 ng/0.5 microl) elevated Fos-ir in the VP to 423% above the contralateral, vehicle-injected side. Bicuculline 52-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 92-95 18663473-6 2008 Likewise, intra-VP NMDA (0.23 microg or 0.45 microg/0.5 microl), dose-dependently increased the number of pallidal neurons expressing Fos-ir by 224 and 526%, respectively. N-Methylaspartate 19-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-137 18663473-7 2008 At higher NMDA doses, the density of Fos-ir neurons was not elevated above control levels. N-Methylaspartate 10-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-40 18663473-9 2008 The mSTN showed a 289% increase in Fos-ir neurons with intra-VP injections of 0.45 microg NMDA, and this response was halved following intra-VP injections of 0.9 microg NMDA. N-Methylaspartate 90-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 18663473-10 2008 Of the 12 other brain regions measured, three showed VP NMDA-induced enhancements in Fos-ir: the frontal cortex, entopeduncular nucleus and substantia nigra pars reticulata, all regions associated with the basal ganglia. N-Methylaspartate 56-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-88 18663473-12 2008 Comparisons of responses to intra-VP NMDA between the hemispheres ipsilateral and contralateral to the lesion revealed that Fos-ir cells in the pedunculopontine nucleus was reduced by 62%, whereas Fos-ir for the basolateral amygdala and STN was reduced by 32 and 42%, respectively. N-Methylaspartate 37-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-127 18485622-0 2008 Morphine and methadone pre-exposures differently modify brain regional Fos protein expression and locomotor activity responses to morphine challenge in the rat. Morphine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 18485622-0 2008 Morphine and methadone pre-exposures differently modify brain regional Fos protein expression and locomotor activity responses to morphine challenge in the rat. Methadone 13-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 18485622-0 2008 Morphine and methadone pre-exposures differently modify brain regional Fos protein expression and locomotor activity responses to morphine challenge in the rat. Morphine 130-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-74 18485622-2 2008 We compared the effects of repeated daily and every-other-day pre-exposure of rats to s.c. morphine and methadone on locomotor activity and CNS neuronal activation (as assessed by Fos immunohistochemistry) responses to s.c. morphine challenge given 2 weeks after the completion of the pretreatment. Morphine 91-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 180-183 18485622-4 2008 Dorsomedial striatum and basolateral amygdaloid nucleus showed robust morphine-induced Fos protein induction that was unaffected by the pretreatments tested. Morphine 70-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-90 18485622-6 2008 Minor Fos responses to morphine were also found in layers IV and VI of the somatosensory cortex and layer VI of the insular cortex of the drug-naive rats; these responses were significantly enhanced both by morphine and methadone pretreatment. Morphine 23-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 6-9 18485622-6 2008 Minor Fos responses to morphine were also found in layers IV and VI of the somatosensory cortex and layer VI of the insular cortex of the drug-naive rats; these responses were significantly enhanced both by morphine and methadone pretreatment. Morphine 207-215 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 6-9 18485622-6 2008 Minor Fos responses to morphine were also found in layers IV and VI of the somatosensory cortex and layer VI of the insular cortex of the drug-naive rats; these responses were significantly enhanced both by morphine and methadone pretreatment. Methadone 220-229 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 6-9 18830027-5 2008 Furthermore, we have shown that manumycin increased the activity of c-Fos in the M-CR3B cells and decreased the activity of NF-kB, while L-NAME reduced the activities of both transcription factors, and accelerated apoptosis of M-CR3B cells. manumycin 32-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 18831893-5 2008 In the VTA, morphine and footshock had an interactive effect on the increase in Fos expression. Morphine 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 18953088-0 2008 Dopamine D1-like receptors agonist SKF 38393 increases cFOS expression in the paraventricular nucleus of the hypothalamus--impact of acute and chronic cocaine. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 35-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-59 18953088-5 2008 We also found that both acute and repeated (once daily for 5 consecutive days) exposure to cocaine (25 mg/kg) attenuated the induction of cFos expression triggered by SKF 38393 when administered 24 hours after single or the last dose of cocaine (25 mg/kg). Cocaine 91-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-142 18953088-5 2008 We also found that both acute and repeated (once daily for 5 consecutive days) exposure to cocaine (25 mg/kg) attenuated the induction of cFos expression triggered by SKF 38393 when administered 24 hours after single or the last dose of cocaine (25 mg/kg). Cocaine 237-244 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 138-142 18495833-1 2008 Intraoral infusions of bitter tastants activate expression of the immediate-early gene c-Fos in neurons located in the medial third of the rostral nucleus of the solitary tract (rNST). bitter tastants 23-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 18539009-0 2008 Region-specific involvement of AMPA/Kainate receptors in Fos protein expression induced by cocaine-conditioned cues. Cocaine 91-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 18634767-8 2008 In contrast, animals that were trained with the light alone (no fear conditioning) and were injected with amphetamine had high levels of c-fos mRNA in the CEAl/c and BSTov. Amphetamine 106-117 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 137-142 18634767-9 2008 Animals that underwent fear conditioning, and were re-exposed to the conditioned stimulus after amphetamine injection had significantly reduced levels of c-fos mRNA in both the BSTov and CEAl/c, compared to the non-conditioned animals. Amphetamine 96-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 19211972-1 2008 The effect of an anxiolytic drug, midazolam, on the expression of c-Fos protein (the product of the immediate early gene, c-fos) in the rat brain was studied in animals that were exposed to the stress of neophobia using the open field test. Midazolam 34-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 19211972-1 2008 The effect of an anxiolytic drug, midazolam, on the expression of c-Fos protein (the product of the immediate early gene, c-fos) in the rat brain was studied in animals that were exposed to the stress of neophobia using the open field test. Midazolam 34-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 19211972-2 2008 Midazolam (0.5 mg/kg, ip) selectively and significantly attenuated the neophobia-induced increase in the number of Fos-like immunoreactive neurons in the dorsomedial part of the prefrontal cortex, but not in the primary motor cortex, the piriform cortex or the amygdalar nuclei. Midazolam 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-118 18539009-1 2008 This study investigated the effects of the AMPA/Kainate receptor antagonist, NBQX, on cue-elicited cocaine-seeking behavior and concomitant changes in Fos protein expression. 2,3-dioxo-6-nitro-7-sulfamoylbenzo(f)quinoxaline 77-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-154 18539009-4 2008 NBQX markedly attenuated cue-elicited cocaine-seeking behavior relative to vehicle pretreatment in the No Extinction group and also decreased cue-elicited Fos protein expression in a region-specific manner in the anterior cingulate and orbitofrontal cortices, basolateral amygdala, nucleus accumbens core, and dorsal caudate-putamen, suggesting involvement of AMPA glutamate systems in specific subregions of the neuronal circuitry activated by cocaine cues. 2,3-dioxo-6-nitro-7-sulfamoylbenzo(f)quinoxaline 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 155-158 18308791-5 2008 0.5 M sodium lactate or saline, in control and panic-prone rats on c-Fos expression in serotonergic neurons within subdivisions of the midbrain/pontine raphe nuclei. Sodium Lactate 6-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 18308791-5 2008 0.5 M sodium lactate or saline, in control and panic-prone rats on c-Fos expression in serotonergic neurons within subdivisions of the midbrain/pontine raphe nuclei. Sodium Chloride 24-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 18308791-7 2008 Lactate increased c-Fos expression in serotonergic neurons located in the ventrolateral part of the dorsal raphe nucleus (DRVL) and ventrolateral periaqueductal gray (VLPAG) of control, but not panic-prone, rats. Lactic Acid 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 17987061-10 2008 Chronic fluoxetine additionally increased c-fos expression in the anterior nucleus accumbens (aAcb) and the piriform cortex (Pir). Fluoxetine 8-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 18588948-6 2008 Two-way ANOVA revealed a significant decrease of c-Fos-immunoreactive (ir) cells expression in all regions of the rostral PAG after both acute and chronic buspirone (acute buspirone (AB) and chronic buspirone (CB), respectively) treatment. Buspirone 155-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 18556369-5 2008 Balloon inflation over a 3 h period following Furo-Cap treatment decreased Fos-ir in the organum vasculosum of the lamina terminalis and the subfornical organ and increased Fos-ir in the lateral parabrachial nucleus and caudal ventrolateral medulla. furo 46-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 18541319-0 2008 The kynurenate analog SZR-72 prevents the nitroglycerol-induced increase of c-fos immunoreactivity in the rat caudal trigeminal nucleus: comparative studies of the effects of SZR-72 and kynurenic acid. Kynurenic Acid 4-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 18541319-0 2008 The kynurenate analog SZR-72 prevents the nitroglycerol-induced increase of c-fos immunoreactivity in the rat caudal trigeminal nucleus: comparative studies of the effects of SZR-72 and kynurenic acid. Nitroglycerin 42-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-81 18541319-1 2008 Administration of nitroglycerol in a migraine model results in an increased number of c-fos-expressing secondary sensory neurons in the caudal trigeminal nucleus. Nitroglycerin 18-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 86-91 18541319-3 2008 Systemic treatment of rats with SZR-72, a newly synthetized kynurenic acid analog, diminished the nitroglycerol-induced increase of c-fos immunoreactivity in the brain stem highly significantly, while treatment with kynurenic acid resulted in a significantly smaller decrease, proving that SZR-72 is much more effective than kynurenic acid. Kynurenic Acid 60-74 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 18541319-3 2008 Systemic treatment of rats with SZR-72, a newly synthetized kynurenic acid analog, diminished the nitroglycerol-induced increase of c-fos immunoreactivity in the brain stem highly significantly, while treatment with kynurenic acid resulted in a significantly smaller decrease, proving that SZR-72 is much more effective than kynurenic acid. Nitroglycerin 98-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-137 18588948-6 2008 Two-way ANOVA revealed a significant decrease of c-Fos-immunoreactive (ir) cells expression in all regions of the rostral PAG after both acute and chronic buspirone (acute buspirone (AB) and chronic buspirone (CB), respectively) treatment. Buspirone 172-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 18588948-6 2008 Two-way ANOVA revealed a significant decrease of c-Fos-immunoreactive (ir) cells expression in all regions of the rostral PAG after both acute and chronic buspirone (acute buspirone (AB) and chronic buspirone (CB), respectively) treatment. Buspirone 172-181 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 18588948-8 2008 Interestingly, c-Fos-ir cells in the dorsomedial periaqueductal gray (dmPAG) column were reduced consistently in both the rostral and caudal PAG in both AB and CB groups. dmpag 70-75 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 15-20 18554809-8 2008 Induction of Fos expression by acute immobilization stress was comparable following CIH in several HPA-modulatory brain regions, including the paraventricular nucleus, bed nucleus of the stria terminalis, and amygdala. cih 84-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-16 19100119-10 2008 The expressions of p-JAK2 and p-STAT3 as well as the expression of AGT mRNA and c-fos mRNA significantly activated by CT-1, which could be inhibited by SIM or Janus Kinase-selective inhibitor AG490. alpha-cyano-(3,4-dihydroxy)-N-benzylcinnamide 192-197 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 18515016-8 2008 The results demonstrated that patterned stimulation elicited a significant increase in c-Fos immunolabeled neurons in layer IV of the contralateral primary visual cortex to the stimulated eye which was completely abolished by cholinergic fibers lesion as well as scopolamine administration. Scopolamine 263-274 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 17976913-6 2008 Either tactile stimulation of the hindpaw ipsilateral to nerve injury, or intraplantar injection of noxious formalin, increased the number of Fos-like immunoreactive profiles. Formaldehyde 108-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 142-145 17976913-8 2008 We found that intrathecal NPY reduced both formalin- and SNI-induced Fos expression. Formaldehyde 43-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 69-72 17976913-9 2008 NPY inhibition of SNI-induced Fos expression was localized to the sural (uninjured) innervation territory, and could be blocked by intrathecal BIBO3304 and BIIE0246. BIBO 3304 143-151 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 17976913-9 2008 NPY inhibition of SNI-induced Fos expression was localized to the sural (uninjured) innervation territory, and could be blocked by intrathecal BIBO3304 and BIIE0246. BIIE 0246 156-164 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-33 18524486-5 2008 The expression of the immediate-early gene product Fos in the dorsal horn ipsilateral to formalin injection was similar between QUIS- and sham-lesioned rats. Formaldehyde 89-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-54 18485423-10 2008 Increased Fos labelling was also observed in the ventral and dorsal aspects of the PAG, a region involved in anxiety-related processes suggesting that this region could be a common neural substrate enlisted during anxiety evoked by dangerous stimuli as well as those elicited by opiate withdrawal. pag 83-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-13 18485423-10 2008 Increased Fos labelling was also observed in the ventral and dorsal aspects of the PAG, a region involved in anxiety-related processes suggesting that this region could be a common neural substrate enlisted during anxiety evoked by dangerous stimuli as well as those elicited by opiate withdrawal. Opiate Alkaloids 279-285 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-13 18587450-6 2008 Similarly, in the pilocarpine-induced status epilepticus (SE) model, BmK IT2 significantly prolonged the latency to onset of the SE, reduced the severity of SE and suppressed hippocampal c-Fos expression during SE. Pilocarpine 18-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 187-192 18403478-3 2008 As adults, neonatal GDX rats showed higher levels of plasma corticosterone and Fos activation in medial parvocellular part of the paraventricular nucleus of the hypothalamus under basal conditions and during 30 min of restraint exposure. GDP-alpha-D-mannuronic acid 20-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 79-82 18554809-10 2008 By contrast, acute Fos induction was enhanced in noradrenergic neurons in the locus coeruleus following CIH exposure. cih 104-107 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 19-22 18188611-5 2008 Significant decreases in c-Fos positive nuclei in the PVN and LC, and enhancement of c-Fos expression in the NA and NTS were induced by NAI. nai 136-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 18188611-5 2008 Significant decreases in c-Fos positive nuclei in the PVN and LC, and enhancement of c-Fos expression in the NA and NTS were induced by NAI. nai 136-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 85-90 18338798-5 2008 Impaired fos mRNA responses to nicotine administration in the celiac ganglia of 6-OHDA-pretreated rats correlated temporally with suppressed expression of functional nicotinic receptors. Nicotine 31-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 18338798-5 2008 Impaired fos mRNA responses to nicotine administration in the celiac ganglia of 6-OHDA-pretreated rats correlated temporally with suppressed expression of functional nicotinic receptors. Oxidopamine 80-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 9-12 18338798-8 2008 The principal neurons in rat celiac ganglia were reflexively activated by 2-deoxy-glucose-induced glucopenia, and the Fos response in the celiac ganglia was markedly inhibited by pretreatment with 6-OHDA. Oxidopamine 197-203 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-121 17851539-5 2008 In the second experiment, c-Fos activation after reinstatement of ethanol seeking induced by discrete cues was compared with the activation pattern of its putative partner (c-Jun) and regulators (extracellular signal-regulated kinases and c-Jun N-terminal kinases). Ethanol 66-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 18270244-10 2008 In vagal capsaicin-treated rats subjected to PVS, Fos expression was significantly decreased in both the solitary tract nucleus and paraventricular nucleus compared with untreated PVS rats. Capsaicin 9-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 17851539-6 2008 Reexposure to ethanol-associated context cues (an extinction session) potentiated c-Fos expression within the basolateral and central amygdala. Ethanol 14-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 17851539-7 2008 Repeated presentation of ethanol-associated discrete cues in an extinction/reinstatement session led to even stronger c-Fos activation in the latter nuclei. Ethanol 25-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 118-123 18397915-5 2008 The present study examined the dose-related impact of DMDC on hypothalamic GnRH neuronal activation indicated by the nuclear presence of the early gene product c-fos. Dimethyldithiocarbamate 54-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 18581599-8 2008 In rats with chronic spinal cord injury by intravesical saline instillation, 82.4+/-10.3% of PGNs in SPN exhibited Fos-immunoreactive (IR). Sodium Chloride 56-62 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-118 18581599-8 2008 In rats with chronic spinal cord injury by intravesical saline instillation, 82.4+/-10.3% of PGNs in SPN exhibited Fos-immunoreactive (IR). pgns 93-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 115-118 18397915-8 2008 A dose-related decline in hypothalamic NE and increase in DA at 2 h after DMDC administration were consistent with a decrease in c-fos-positive GnRH neurons, with an almost complete absence of c-fos at the two highest doses. Dimethyldithiocarbamate 74-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 129-134 18576208-0 2008 Patterns of FOS expression in the spinal cord and periaqueductal grey matter of 6OHDA-lesioned rats. Oxidopamine 80-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-15 18397915-9 2008 The effects correlated well with a suppression of the surge, although the percentage decrease in c-fos neurons at 7.1 mg/kg only attenuated the surge peak, not the overall amount of circulating LH. Luteinizing Hormone 194-196 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 18499630-8 2008 RESULTS: Exposure to 75% N(2)O increased c-Fos expression in tyrosine hydroxylase-positive cells in the ventral tegmental area. Nitrous Oxide 25-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-46 18576208-3 2008 This study explores the patterns of Fos expression (a well-known marker for changes in cell activity) in the spinal cord and periaqueductal grey matter (PaG), two major sensory (nociceptive) centers, of hemiParkinsonian rats. pag 153-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-39 18576208-7 2008 In the PaG, there were many more Fos(+) cells in the 6OHDA-lesioned than in the Control group, in both the stimulation and, in particular, the non-stimulation cases. pag 7-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 18576208-7 2008 In the PaG, there were many more Fos(+) cells in the 6OHDA-lesioned than in the Control group, in both the stimulation and, in particular, the non-stimulation cases. Oxidopamine 53-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-36 18576208-8 2008 In the spinal cord, there were also more Fos(+) cells in the 6OHDA-lesioned than in the Control group, but in the stimulation cases only. Oxidopamine 61-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 41-44 18576208-9 2008 Overall, the results show distinct changes in Fos expression in the spinal cord and PaG of 6OHDA-lesioned rats, suggesting a substrate for some of the abnormal sensory (nociceptive) circuits that may be evident in parkinsonian cases. Oxidopamine 91-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 18457920-7 2008 However, these signals of c-fos, c-jun, and nur77 by hypoxia were reduced significantly by naloxone. Naloxone 91-99 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 18457920-9 2008 The induction of c-fos, c-jun, nur77, and zif268 by hypoxia was inhibited by naloxone (0.1 microM) and MAPK inhibitors (10 microM of U0126, D98059, SB203580). Naloxone 77-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 18457920-9 2008 The induction of c-fos, c-jun, nur77, and zif268 by hypoxia was inhibited by naloxone (0.1 microM) and MAPK inhibitors (10 microM of U0126, D98059, SB203580). U 0126 133-138 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 18457920-9 2008 The induction of c-fos, c-jun, nur77, and zif268 by hypoxia was inhibited by naloxone (0.1 microM) and MAPK inhibitors (10 microM of U0126, D98059, SB203580). SB 203580 148-156 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 18457920-11 2008 Thus, the present studies demonstrated that OGD induced IEGs including c-fos, c-jun, nur77, and zif268 and MAPK signaling pathways were regulated differently by naloxone. Naloxone 161-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 18485334-6 2008 In the hippocampus, a single corticosterone dose (10 mg/kg, s.c.) caused a widespread and transient reduction of fosB mRNA after 0.8 h, whereas changes in both c-fos and fra-1 mRNA levels were restricted to the dentate gyrus region. Corticosterone 29-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 18495107-7 2008 Inhibition of dopamine release by isoliquiritigenin resulted in attenuation of the expression of c-Fos, an immediately early gene induced by cocaine. Dopamine 14-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 18495107-7 2008 Inhibition of dopamine release by isoliquiritigenin resulted in attenuation of the expression of c-Fos, an immediately early gene induced by cocaine. isoliquiritigenin 34-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 18495107-7 2008 Inhibition of dopamine release by isoliquiritigenin resulted in attenuation of the expression of c-Fos, an immediately early gene induced by cocaine. Cocaine 141-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 97-102 17999187-6 2008 Toluidine blue improved method (TBI) and immunohistochemistry have also been involved in observations of mucous mast cells in the colon, change of c-fos positive cells, and secretion of SP, SPR, VIP, VIPR in the local colon. Tolonium Chloride 0-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-152 17999187-6 2008 Toluidine blue improved method (TBI) and immunohistochemistry have also been involved in observations of mucous mast cells in the colon, change of c-fos positive cells, and secretion of SP, SPR, VIP, VIPR in the local colon. tbi 32-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-152 18172855-0 2008 Lysophosphatidic acid-induced c-fos up-regulation involves cyclic AMP response element-binding protein activated by mitogen- and stress-activated protein kinase-1. lysophosphatidic acid 0-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 18588535-8 2008 Nicotine-exposed adolescents did not self-administer the low dose of cocaine, but, at the higher dose, exhibited significantly greater cocaine intake and c-fos mRNA expression in nucleus accumbens than did controls. Nicotine 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 154-159 18172855-3 2008 Previously, LPA has been shown to stimulate c-fos mRNA expression in Rat-2 fibroblast cells via a serum response element binding protein (SRF). lysophosphatidic acid 12-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-49 18540883-5 2008 Moreover, exogenous replacement of the main metabolite of COX-2 activity, PGE(2), was sufficient to restore LTP induction and for normal downstream signalling to ensue, namely extracellular signalling-regulated kinase (ERK)-phosphorylation and c-FOS expression. Prostaglandins E 74-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 244-249 18172855-0 2008 Lysophosphatidic acid-induced c-fos up-regulation involves cyclic AMP response element-binding protein activated by mitogen- and stress-activated protein kinase-1. Cyclic AMP 59-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 18172855-4 2008 However, involvement of CREB in LPA-stimulated c-fos gene expression is not elucidated yet. lysophosphatidic acid 32-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-52 18172855-5 2008 To investigate the CREB-mediated c-fos activation by LPA, various c-fos promoter-reporter constructs containing wild-type and mutated SRE and CRE were tested for their inducibility by LPA in transient transfection assays. lysophosphatidic acid 53-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 18314190-0 2008 Cocaine administration increases the fraction of CART cells in the rat nucleus accumbens that co-immunostain for c-Fos. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 113-118 18172855-7 2008 A dominant negative CREB significantly down-regulated the LPA-stimulated c-fos promoter activation. lysophosphatidic acid 58-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 18172855-8 2008 Chromatin immunoprecipitation assay revealed that LPA induced an increased binding of phosphorylated CREB and CREB-binding protein (CBP) to the CRE region of the endogenous c-fos promoter. lysophosphatidic acid 50-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 173-178 18172855-9 2008 Immunoblot analyses with various pharmacological inhibitors further showed that LPA induces up-regulation of c-fos mRNA level by activation of ERK, p38 MAPK, and MSK1. lysophosphatidic acid 80-83 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 18383504-6 2008 Consistent with its behavioral effects, ketamine induced Fos expression in cholinergic, monoaminergic, and orexinergic arousal systems and completely suppressed Fos immunoreactivity in the sleep-promoting ventrolateral preoptic nucleus (VLPO). Ketamine 40-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 18383504-6 2008 Consistent with its behavioral effects, ketamine induced Fos expression in cholinergic, monoaminergic, and orexinergic arousal systems and completely suppressed Fos immunoreactivity in the sleep-promoting ventrolateral preoptic nucleus (VLPO). Ketamine 40-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 161-164 18583263-1 2008 OBJECTIVE: To investigate the mechanism of rosiglitazone (RSG, the activator of peroxisome proliferators activated receptor lambda) for inhibiting endothelin-1 (ET-1)-induced neonatal rat cardiac myocyte hypertrophy and the role of protein kinase C (PKC) and c-fos. Rosiglitazone 43-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 259-264 18583263-1 2008 OBJECTIVE: To investigate the mechanism of rosiglitazone (RSG, the activator of peroxisome proliferators activated receptor lambda) for inhibiting endothelin-1 (ET-1)-induced neonatal rat cardiac myocyte hypertrophy and the role of protein kinase C (PKC) and c-fos. Rosiglitazone 58-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 259-264 18417125-6 2008 Measurements of amphetamine-induced striatal c-fos expression, as well as behavior results gathered when animals were under the influence of apomorphine or haloperidol, indicate that this potential reorganization may require non-dopaminergic neural plasticity. Amphetamine 16-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 18583263-7 2008 RSG inhibited protein synthesis enhancement and increased (3)H-leucine incorporation induced by ET-1 and PMA, and antagonized the effects of ET-1 and PMA in promoting PKC activity and c-fos protein expression in the myocytes. Rosiglitazone 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 184-189 18583263-7 2008 RSG inhibited protein synthesis enhancement and increased (3)H-leucine incorporation induced by ET-1 and PMA, and antagonized the effects of ET-1 and PMA in promoting PKC activity and c-fos protein expression in the myocytes. Tetradecanoylphorbol Acetate 150-153 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 184-189 18583263-8 2008 CONCLUSION: The inhibitory effect of RSG on ET-1- or PMA-induced myocyte hypertrophy is associated with PKC-c-fos pathway. Rosiglitazone 37-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-113 18396266-7 2008 In contrast, although peak levels of dopamine were unaffected by infusion rate, dopamine levels increased more rapidly when cocaine was administered over 5 versus 100 s. Moreover, c-fos mRNA expression in the region of the striatum sampled was greater when cocaine was administered rapidly than when given slowly. Dopamine 80-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 180-185 18314190-2 2008 There was about a 45% increase in the fraction of cells that stained for both CART and c-Fos after administration of cocaine, but there was no change in the fraction after administration of saline. Cocaine 117-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 87-92 18374904-9 2008 In situ hybridization confirmed that c-Fos and Fra-2 mRNA expression was increased in the CeA after LiCl and sucrose/LiCl treatment. cea 90-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 18374904-9 2008 In situ hybridization confirmed that c-Fos and Fra-2 mRNA expression was increased in the CeA after LiCl and sucrose/LiCl treatment. Lithium Chloride 100-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 18374904-9 2008 In situ hybridization confirmed that c-Fos and Fra-2 mRNA expression was increased in the CeA after LiCl and sucrose/LiCl treatment. Sucrose 109-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 18374904-9 2008 In situ hybridization confirmed that c-Fos and Fra-2 mRNA expression was increased in the CeA after LiCl and sucrose/LiCl treatment. Lithium Chloride 117-121 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 37-42 18187539-4 2008 Intracerebroventricular administration of the MAPK inhibitor, PD98059, prevented LPS-induced ERK1/2 phosphorylation, c-fos activation, and the increase of CRH gene expression in the PVN but had no effect on c-fos activation in brainstem A2-C1/C2 regions. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 62-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 117-122 18187539-4 2008 Intracerebroventricular administration of the MAPK inhibitor, PD98059, prevented LPS-induced ERK1/2 phosphorylation, c-fos activation, and the increase of CRH gene expression in the PVN but had no effect on c-fos activation in brainstem A2-C1/C2 regions. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 62-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 207-212 18513318-6 2008 In rats receiving vehicle injections, swim, a behavior known to influence 5-HT neurotransmission, increased the number of Fos-containing 5-HT cells only in the caudal third of DR. Administration of WAY-100635 preceding a swim did not change the amount of Fos in the caudal DR, but increased the number of Fos-containing 5-HT cells in the rostral DR, lateral wings of the DR, and MR. N-(2-(4-(2-methoxyphenyl)-1-piperazinyl)ethyl)-N-(2-pyridinyl)cyclohexanecarboxamide 198-208 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-125 18092361-9 2008 Significant amounts of NADPH-d-stained neurons in the PVN of water-deprived rats (67-68% in both the mP and the pM) exhibited c-Fos-ir. Water 61-66 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 126-131 18092361-10 2008 NADPH-d/c-Fos neurons in the pM-PVN were increased by water deprivation but not changed by rehydration. NADP 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-13 18092361-10 2008 NADPH-d/c-Fos neurons in the pM-PVN were increased by water deprivation but not changed by rehydration. Water 54-59 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 8-13 18347780-10 2008 The c-fos response to amphetamine in the accumbens core was augmented in amphetamine-pretreated animals with a shift in the distribution of optical density, while no effect of sensitization was seen in the nucleus accumbens shell or prefrontal cortex. Amphetamine 22-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 18347780-10 2008 The c-fos response to amphetamine in the accumbens core was augmented in amphetamine-pretreated animals with a shift in the distribution of optical density, while no effect of sensitization was seen in the nucleus accumbens shell or prefrontal cortex. Amphetamine 73-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 18438645-6 2008 Moreover, at the neural level, DMI treatment led to a decrease in FST-induced c-fos messenger RNA levels in medial prefrontal cortex (PFC) and paraventricular nucleus of the hypothalamus (PVN) in LR but not HR animals. Desipramine 31-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 18255166-1 2008 We determined if cutaneous hyperalgesia and pain-induced c-Fos overexpression in the spinal cord produced by repeated forced swimming (FS) stress in the rat were related to changes in GABA neurotransmission by studying spinal release of GABA and the effect of positive modulation of GABA-A receptors with diazepam. gamma-Aminobutyric Acid 184-188 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 18255166-8 2008 In FS rats, diazepam did not have effect on GABA release but reduced pain scores and overexpression of c-Fos whereas flumazenil (0.1 mg/kg, i.p. Diazepam 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 18396266-0 2008 The rate of intravenous cocaine administration alters c-fos mRNA expression and the temporal dynamics of dopamine, but not glutamate, overflow in the striatum. Cocaine 24-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-59 18478050-4 2008 To investigate this question, we examined where the dopamine D1-like receptor agonist, SKF 38393, altered Fos expression via estrogen receptor activation. 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine 87-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 18395195-11 2008 c-fos immunoreactivity in brain sections of kindled rats with CD displayed a striking increase by convulsive PTZ challenge. Pentylenetetrazole 109-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 18460697-10 2008 Immunohistochemical studies demonstrated choline acetyltransferase immunoreactivity in response to L-Glu microinjection into the rostral DMV in 88% of c-Fos-positive intragastric myenteric neurons. Glutamic Acid 99-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-156 18460697-10 2008 Immunohistochemical studies demonstrated choline acetyltransferase immunoreactivity in response to L-Glu microinjection into the rostral DMV in 88% of c-Fos-positive intragastric myenteric neurons. (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 137-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-156 18460697-11 2008 Microinjection of L-Glu into the caudal DMV evoked expression of nitric oxide (NO) synthase and VIP immunoreactivity in 81 and 39%, respectively, of all c-Fos-positive intragastric myenteric neurons. Glutamic Acid 18-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-158 18460697-11 2008 Microinjection of L-Glu into the caudal DMV evoked expression of nitric oxide (NO) synthase and VIP immunoreactivity in 81 and 39%, respectively, of all c-Fos-positive intragastric myenteric neurons. (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 40-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-158 18460697-11 2008 Microinjection of L-Glu into the caudal DMV evoked expression of nitric oxide (NO) synthase and VIP immunoreactivity in 81 and 39%, respectively, of all c-Fos-positive intragastric myenteric neurons. Nitric Oxide 65-77 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-158 18374904-13 2008 Fra-2 and c-Fos were examined in response to sucrose/LiCl in rats with prior familiarity with sucrose compared to rats without prior exposure to sucrose. Sucrose 45-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-15 18374904-13 2008 Fra-2 and c-Fos were examined in response to sucrose/LiCl in rats with prior familiarity with sucrose compared to rats without prior exposure to sucrose. Lithium Chloride 53-57 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 10-15 18396266-7 2008 In contrast, although peak levels of dopamine were unaffected by infusion rate, dopamine levels increased more rapidly when cocaine was administered over 5 versus 100 s. Moreover, c-fos mRNA expression in the region of the striatum sampled was greater when cocaine was administered rapidly than when given slowly. Cocaine 124-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 180-185 18396266-7 2008 In contrast, although peak levels of dopamine were unaffected by infusion rate, dopamine levels increased more rapidly when cocaine was administered over 5 versus 100 s. Moreover, c-fos mRNA expression in the region of the striatum sampled was greater when cocaine was administered rapidly than when given slowly. Cocaine 257-264 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 180-185 18400210-7 2008 The acute effect of the 5-HT2A/2C receptor agonist DOI and glutamate administration at various circadian times on c-fos and Per1 expressions was assessed. Glutamic Acid 59-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-119 18400210-8 2008 Glutamate induced the expression of c-fos at CT6, CT16 and CT22, but induced Per1 expression at CT16 only. Glutamic Acid 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 18358631-0 2008 Acute ethanol induces Fos in GABAergic and non-GABAergic forebrain neurons: a double-labeling study in the medial prefrontal cortex and extended amygdala. Ethanol 6-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-25 18425310-0 2008 H(2)S protects myocardium against ischemia/reperfusion injury and its effect on c-Fos protein expression in rats. Hydrogen Sulfide 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-85 18425310-13 2008 NaHS at 14 mumol/kg body weight markedly alleviated the injury in morphological changes and decreased c-Fos protein expression in myocardial tissue compared with that in I/R group (0.20+-0.06 vs 0.32+-0.10, P<0.05). sodium bisulfide 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 18425310-14 2008 These results suggest that H(2)S protects myocardium against I/R injury and this protective effect may be related to the down-regulation of c-Fos protein expression. Hydrogen Sulfide 27-32 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 18358631-5 2008 Intragastric water administration increased the number of Fos-immunoreactive cells in the infralimbic cortex and lateral part of the central nucleus of the amygdala compared with the naive group. Water 13-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 18358631-6 2008 Ethanol administration increased the number of Fos-immunoreactive cells in the infralimbic (+57.5%) and prelimbic (+105.3%) cortices, nucleus accumbens shell region (+88.2%), medial part of the central nucleus of the amygdala (+160%), and lateral part of the bed nucleus of the stria terminalis (+198.8%) compared with the water-treated group. Ethanol 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 18358631-6 2008 Ethanol administration increased the number of Fos-immunoreactive cells in the infralimbic (+57.5%) and prelimbic (+105.3%) cortices, nucleus accumbens shell region (+88.2%), medial part of the central nucleus of the amygdala (+160%), and lateral part of the bed nucleus of the stria terminalis (+198.8%) compared with the water-treated group. Water 323-328 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 18358631-7 2008 In the nucleus accumbens shell region, central nucleus of the amygdala, and bed nucleus of the stria terminalis, more than 80% of Fos-immunoreactive neurons were GABAergic after ethanol administration. Ethanol 178-185 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 130-133 17902169-9 2008 S1P-stimulated AP-1 subunits (c-Jun and c-Fos mRNA) expression was attenuated by U0126 and wortmannin, suggesting that MEK and PI3K/ERK cascade linking to AP-1 was involved in EGFR expression. U 0126 81-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 17902169-9 2008 S1P-stimulated AP-1 subunits (c-Jun and c-Fos mRNA) expression was attenuated by U0126 and wortmannin, suggesting that MEK and PI3K/ERK cascade linking to AP-1 was involved in EGFR expression. Wortmannin 91-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 18349210-0 2008 Suppression of noxious-induced c-fos expression in the rat lumbar spinal cord by isoflurane alone or combined with fentanyl. Isoflurane 81-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 18329635-6 2008 Fos-ir positive neurons were mainly localized within the ventral part of the DMH. Dimenhydrinate 77-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 18329635-8 2008 Additional double-labelling with anti-Fos and anti-AgRP revealed that Fos positive neurons in the DMH were encircled by a network of AgRP-ir positive fibers. Dimenhydrinate 98-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 18329635-8 2008 Additional double-labelling with anti-Fos and anti-AgRP revealed that Fos positive neurons in the DMH were encircled by a network of AgRP-ir positive fibers. Dimenhydrinate 98-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 18313647-4 2008 The suppressive effects were detected at 2, 4, 6, 8, and 12 h after ACPD injection, but returned to the control level at 24 h. A remarkable c-fos expression was found in the lesioned side of GP, subthalamic nucleus (STN), and substantia nigra pars reticulata (SNr) of rats that received the ACPD or DHPG injection, compared to rats treated with L-AP-4 or phosphate buffer-injection. 1-aminocyclopentane-1,3-dicarboxylic acid 68-72 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 18313647-4 2008 The suppressive effects were detected at 2, 4, 6, 8, and 12 h after ACPD injection, but returned to the control level at 24 h. A remarkable c-fos expression was found in the lesioned side of GP, subthalamic nucleus (STN), and substantia nigra pars reticulata (SNr) of rats that received the ACPD or DHPG injection, compared to rats treated with L-AP-4 or phosphate buffer-injection. 1-aminocyclopentane-1,3-dicarboxylic acid 291-295 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 18313647-4 2008 The suppressive effects were detected at 2, 4, 6, 8, and 12 h after ACPD injection, but returned to the control level at 24 h. A remarkable c-fos expression was found in the lesioned side of GP, subthalamic nucleus (STN), and substantia nigra pars reticulata (SNr) of rats that received the ACPD or DHPG injection, compared to rats treated with L-AP-4 or phosphate buffer-injection. 3,5-dihydroxyphenylglycine 299-303 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 18313647-4 2008 The suppressive effects were detected at 2, 4, 6, 8, and 12 h after ACPD injection, but returned to the control level at 24 h. A remarkable c-fos expression was found in the lesioned side of GP, subthalamic nucleus (STN), and substantia nigra pars reticulata (SNr) of rats that received the ACPD or DHPG injection, compared to rats treated with L-AP-4 or phosphate buffer-injection. 2-amino-4-phosphono-propinate 345-351 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 18313647-4 2008 The suppressive effects were detected at 2, 4, 6, 8, and 12 h after ACPD injection, but returned to the control level at 24 h. A remarkable c-fos expression was found in the lesioned side of GP, subthalamic nucleus (STN), and substantia nigra pars reticulata (SNr) of rats that received the ACPD or DHPG injection, compared to rats treated with L-AP-4 or phosphate buffer-injection. Phosphates 355-364 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 18234743-5 2008 Rats injected with CTb in the insular cortex and stimulated with hypertonic saline had increased numbers of Fos/CTb double-positive neurons in the paraventricular, rhomboid, and reuniens thalamic nuclei, whereas those rats injected with CTb in the cingulate cortex and challenged with hypertonic saline had increased numbers of Fos/CTb double-positive neurons in the medial part of the mediodorsal, interanteromedial, anteromedial, and ventrolateral part of the laterodorsal thalamic nuclei. Sodium Chloride 76-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 18234743-5 2008 Rats injected with CTb in the insular cortex and stimulated with hypertonic saline had increased numbers of Fos/CTb double-positive neurons in the paraventricular, rhomboid, and reuniens thalamic nuclei, whereas those rats injected with CTb in the cingulate cortex and challenged with hypertonic saline had increased numbers of Fos/CTb double-positive neurons in the medial part of the mediodorsal, interanteromedial, anteromedial, and ventrolateral part of the laterodorsal thalamic nuclei. Sodium Chloride 76-82 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 328-331 18234743-6 2008 Rats injected with CTb in the dorsal midline of the thalamus and challenged with hypertonic saline had increased numbers of Fos/CTb double-positive neurons within the organum vasculosum of the lamina terminalis (OVLT), median preoptic nucleus, and insular cortex but not the subfornical organ. Sodium Chloride 92-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 124-127 18412612-4 2008 induced expression of the marker genes c-fos and egr-1 in brain regions associated with both rewarding and stressful ethanol actions. Ethanol 117-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 18412612-5 2008 Under non-dependent conditions, ethanol-induced c-fos expression was generally not affected by MEK inhibition, with the exception of the medial amygdala (MeA). Ethanol 32-39 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 18412612-6 2008 In contrast, following a history of dependence, a markedly suppressed c-fos response to acute ethanol was found in the medial pre-frontal/orbitofrontal cortex (OFC), nucleus accumbens shell (AcbSh) and paraventricular nucleus (PVN). Ethanol 94-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 18412617-4 2008 injection of a ghrelin mimetic, GHRP-6, during the daytime activated neurons in the vmARC and reduced the normal endogenous daytime Fos expression in the SCN. Ghrelin 15-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 132-135 17848208-1 2008 OBJECTIVE: In an effort to understand cell activity patterns and sensorimotor integration in Parkinson"s disease, we have explored the expression of the Fos protein in the subthalamus after sensory (nociceptive) stimulation of hemiparkinsonian Sprague-Dawley rats [6-hydroxydopamine [6OHDA]-lesioned]. Oxidopamine 265-282 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-156 17848208-1 2008 OBJECTIVE: In an effort to understand cell activity patterns and sensorimotor integration in Parkinson"s disease, we have explored the expression of the Fos protein in the subthalamus after sensory (nociceptive) stimulation of hemiparkinsonian Sprague-Dawley rats [6-hydroxydopamine [6OHDA]-lesioned]. Oxidopamine 284-289 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 153-156 17848208-7 2008 However, in the cases that had 6OHDA-lesions combined with mechanical stimulation, there were many Fos+ cells within the subthalamus of both sides, particularly on the ipsilateral side. Oxidopamine 31-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 21063301-2 2008 METHODS: Expression of the immediate-early gene c-fos and nitric oxide containing neurons one hour after unilateral formalin injection to the dorsal hind paw was investigated in rat lumbar spinal cord, using fos immunohistochemistry and NADPH-d histochemical techniques. Formaldehyde 116-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 21063301-2 2008 METHODS: Expression of the immediate-early gene c-fos and nitric oxide containing neurons one hour after unilateral formalin injection to the dorsal hind paw was investigated in rat lumbar spinal cord, using fos immunohistochemistry and NADPH-d histochemical techniques. Formaldehyde 116-124 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 50-53 21063301-2 2008 METHODS: Expression of the immediate-early gene c-fos and nitric oxide containing neurons one hour after unilateral formalin injection to the dorsal hind paw was investigated in rat lumbar spinal cord, using fos immunohistochemistry and NADPH-d histochemical techniques. NADP 237-242 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 21063301-5 2008 Approximately 20% of fos-immunoreactive neurons were NADPH-d positive ipsilateral to the formalin injection, whereas no double labeling was observed in the contralateral side. NADP 53-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 21063301-5 2008 Approximately 20% of fos-immunoreactive neurons were NADPH-d positive ipsilateral to the formalin injection, whereas no double labeling was observed in the contralateral side. Deuterium 59-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 21063301-5 2008 Approximately 20% of fos-immunoreactive neurons were NADPH-d positive ipsilateral to the formalin injection, whereas no double labeling was observed in the contralateral side. Formaldehyde 89-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 21-24 21063301-6 2008 Also, a close relation of NADPH-d positive processes with fos-immunoreactive nuclei were also observed. NADP 26-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-61 18080115-0 2008 Repeated amphetamine administration induces Fos in prefrontal cortical neurons that project to the lateral hypothalamus but not the nucleus accumbens or basolateral amygdala. Amphetamine 9-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 44-47 18080115-4 2008 MATERIALS AND METHODS: Using retrograde labeling techniques, Fos activation was evaluated in the predominant projection pathways of the mPFC of sensitized rats after a challenge injection of AMPH. Amphetamine 191-195 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 18080115-5 2008 RESULTS: There was a significant increase in Fos-immunoreactive cells in the mPFC, nucleus accumbens (NAc), basolateral amygdala (BLA), and lateral hypothalamus (LH) of rats treated repeatedly with AMPH when compared to vehicle-treated controls. Amphetamine 198-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 18080115-6 2008 The mPFC pyramidal neurons that project to the LH but not the NAc or BLA show a significant induction of Fos after repeated AMPH treatment. Amphetamine 124-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-108 18262520-2 2008 The objective of the present study was to identify the cell types in the prefrontal cortex in which lysergic acid diethylamide (d-LSD) induces c-Fos expression. Lysergic Acid Diethylamide 100-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-148 18262520-2 2008 The objective of the present study was to identify the cell types in the prefrontal cortex in which lysergic acid diethylamide (d-LSD) induces c-Fos expression. Lysergic Acid Diethylamide 128-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 143-148 18619357-11 2008 If tanshinone II or valsartan was added to the culture medium before Ang II, both of them could inhibit the increase of c-fos, c-jun mRNA expression (P < 0.01), cardiomyocyte protein synthesis rate (P < 0.01), and cardiomyocyte size (P < 0.05) induced by Ang II. tanshinone 3-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 18619357-11 2008 If tanshinone II or valsartan was added to the culture medium before Ang II, both of them could inhibit the increase of c-fos, c-jun mRNA expression (P < 0.01), cardiomyocyte protein synthesis rate (P < 0.01), and cardiomyocyte size (P < 0.05) induced by Ang II. Valsartan 20-29 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 120-125 17434215-0 2008 D-ribose improves cardiac contractility and hemodynamics, and reduces expression of c-fos in the hippocampus during sustained slow ventricular tachycardia in rats. Ribose 0-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-89 17434215-14 2008 CONCLUSION: d-ribose improves hemodynamic parameters, cardiac contractility and prevents the activation of pro-apoptotic c-fos, demonstrating a neuroprotective effect of d-ribose during slow VT. Ribose 12-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-126 17434215-14 2008 CONCLUSION: d-ribose improves hemodynamic parameters, cardiac contractility and prevents the activation of pro-apoptotic c-fos, demonstrating a neuroprotective effect of d-ribose during slow VT. Ribose 170-178 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-126 18282557-4 2008 Intracerebroventricular injection of pilocarpine produced similar changes in the expression of c-Fos immunoreactivity. Pilocarpine 37-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 95-100 18282559-2 2008 Here, we examined the effect of disinhibition of the DMH by unilateral microinjection of bicuculline methiodide (BMI) on Fos expression in selected regions of the brain that have been implicated in anxiety and responses to stress and fever in rats. 1,2-Dimethylhydrazine 53-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-124 18282559-2 2008 Here, we examined the effect of disinhibition of the DMH by unilateral microinjection of bicuculline methiodide (BMI) on Fos expression in selected regions of the brain that have been implicated in anxiety and responses to stress and fever in rats. bicuculline methiodide 89-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-124 18282559-2 2008 Here, we examined the effect of disinhibition of the DMH by unilateral microinjection of bicuculline methiodide (BMI) on Fos expression in selected regions of the brain that have been implicated in anxiety and responses to stress and fever in rats. bicuculline methiodide 113-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 121-124 18282559-3 2008 Disinhibition of the DMH resulted in dramatic increases in local Fos expression and also increased the numbers of Fos-positive neurons in the lateral septal nucleus and in both the parvocellular and magnocellular subdivisions of the paraventricular nucleus, with greater increases ipsilateral to the injection site in the DMH. 1,2-Dimethylhydrazine 21-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-68 18282559-3 2008 Disinhibition of the DMH resulted in dramatic increases in local Fos expression and also increased the numbers of Fos-positive neurons in the lateral septal nucleus and in both the parvocellular and magnocellular subdivisions of the paraventricular nucleus, with greater increases ipsilateral to the injection site in the DMH. 1,2-Dimethylhydrazine 21-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 114-117 18282559-5 2008 In the brainstem, disinhibition of the DMH increased Fos expression in the nucleus tractus solitarius and the ventrolateral medulla bilaterally with greater increases again ipsilateral to the site of the microinjection, and also in the midline rostral raphe pallidus. 1,2-Dimethylhydrazine 39-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 53-56 18282559-6 2008 Thus, disinhibition of neurons in the DMH in conscious rats results in increases in Fos expression in selected forebrain and brainstem regions that have been implicated in stress-induced physiological changes, anxiety, and experimental fever. 1,2-Dimethylhydrazine 38-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 84-87 18160143-9 2008 The c-Fos IR in the nucleus accumbens was altered in a sexually dimorphic manner in the ethanol-exposed group. Ethanol 88-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-9 18280640-6 2008 The DNA binding activity of AP-1 transcription factors and the expression of c-Fos and Fra-2 proteins, were all enhanced when the cAMP and IL-1 signalling pathways were both stimulated. Cyclic AMP 130-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 18155849-0 2008 Fos expression in pontomedullary catecholaminergic cells following rapid eye movement sleep-like episodes elicited by pontine carbachol in urethane-anesthetized rats. Carbachol 126-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 18155849-0 2008 Fos expression in pontomedullary catecholaminergic cells following rapid eye movement sleep-like episodes elicited by pontine carbachol in urethane-anesthetized rats. Urethane 139-147 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 18155849-5 2008 The percentage of Fos-positive TH cells was negatively correlated with the cumulative duration of REMS-like episodes induced during 140 min prior to brain harvesting in the A7 and rostral A5 groups bilaterally (P < 0.01 for both), and in SubC neurons on the side opposite to carbachol injection (P < 0.05). Carbachol 278-287 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 18248910-6 2008 c-Fos immunohistochemistry revealed that DMH CCK increased the number of c-Fos positive cells in the paraventricular nucleus (PVN), arcuate nucleus, suprachiasmatic nucleus and retrochiasmatic area as well as in the contralateral DMH. 1,2-Dimethylhydrazine 41-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 18248910-6 2008 c-Fos immunohistochemistry revealed that DMH CCK increased the number of c-Fos positive cells in the paraventricular nucleus (PVN), arcuate nucleus, suprachiasmatic nucleus and retrochiasmatic area as well as in the contralateral DMH. 1,2-Dimethylhydrazine 41-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 18307678-4 2008 Indeed, the alpha(1)-adrenoceptor antagonist alfuzosin inhibits expression of the oncogene c-fos- a marker of nociceptive pathway activation - evoked by cyclophosphamide in rats. alfuzosin 45-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 18307678-4 2008 Indeed, the alpha(1)-adrenoceptor antagonist alfuzosin inhibits expression of the oncogene c-fos- a marker of nociceptive pathway activation - evoked by cyclophosphamide in rats. Cyclophosphamide 153-169 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-96 18310906-2 2008 It has been shown that repeated injections of cocaine produce an increase in locomotor activity, the expression of the immediate-early gene, c-fos, and the release of dopamine (DA) in the nucleus accumbens (NAc), which is one of the main dopaminergic terminal areas. Cocaine 46-53 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 141-146 18310906-8 2008 significantly inhibited the repeated cocaine-induced increase in locomotor activity as well as the c-Fos expression in the core and shell in a dose-dependent manner. Cocaine 37-44 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 17763940-6 2008 Inhibition of ERK1/2 by PD98059 attenuated Ang II-induced c-fos and c-myc mRNA expression (by 75% and 100%, respectively) but was ineffective in preventing Ang II induction of c-jun. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 24-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 58-63 17950587-4 2008 Thunberginol B inhibited up-regulated genes of all cytokines, and thunberginols A and B (30 microM) inhibited the phosphorylation of c-jun and expression of c-fos mRNA and phosphorylation of extracellular signal-regulated kinases (ERK1/2). thunberginol B 0-14 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-162 17950587-4 2008 Thunberginol B inhibited up-regulated genes of all cytokines, and thunberginols A and B (30 microM) inhibited the phosphorylation of c-jun and expression of c-fos mRNA and phosphorylation of extracellular signal-regulated kinases (ERK1/2). thunberginols 66-79 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 157-162 18311059-5 2008 In the present study, we examined the influence of c-Fos expression in the amygdala in acquisition of conditioned place aversion (CPA) induced by naloxone-precipitated withdrawal from a single morphine exposure 24 h earlier. cpa 130-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 18311059-5 2008 In the present study, we examined the influence of c-Fos expression in the amygdala in acquisition of conditioned place aversion (CPA) induced by naloxone-precipitated withdrawal from a single morphine exposure 24 h earlier. Naloxone 146-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 18311059-7 2008 Findings showed that CeA displayed significant increase in c-Fos expression in the acquisition of CPA. cpa 98-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 59-64 18536377-0 2008 [Effects of beta-asarone on expression of c-fos in kindling epilepsy rat brain]. asarone 12-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 18536377-1 2008 OBJECTIVE: To study the effects of beta-asarone on expression of immediately early gene c-fos in kindling epilepsy rat brain. asarone 35-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 18536377-5 2008 RESULT: Beta-asarone could significantly increase the expression of c-fos in kindling epilepsy rat brain, and show its quantity-effect relation. asarone 8-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 18536377-6 2008 The expression of c-fos in hippocampus was (1139.45 +/- 155.56), (1109.56 +/- 134.03), (1103.73 +/- 235.82) CNT x mm2 in beta-asarone groups, 920.54 +/- 203.20 in model control group, and 1106.26 +/- 186.24 in difetoin group, respectively. asarone 121-133 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 18536377-7 2008 The number of c-fos positive cell was 87.1 +/- 2.2, 76.3 +/- 1.3 and 59.9 +/- 1.3 in beta-asarone groups, 39.3 +/- 2.6 in model control group, and 95.2 +/- 1.1 in difetoin group, respectively. asarone 85-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 18536377-8 2008 CONCLUSION: Beta-asarone can obviously increase the expression of c-fos in epilepsy rat brain. asarone 12-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-71 18343574-0 2008 Expression of Fos protein in brainstem after application of l-menthol to the rat nasal mucosa. Menthol 60-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-17 18343574-3 2008 In the present study, we examined the expression of Fos protein in rat brainstem neurons after nasal application of l-menthol, which is known to activate cold-sensitive nasal receptors. Menthol 116-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-55 18343574-5 2008 Furthermore, a significantly higher density of Fos-immunoreactive cells was observed in the SPV and KF in the l-menthol rats than in the controls. Menthol 110-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 18343574-6 2008 In the SPV, the density of Fos-immunoreactive cells was highest at approximately 0.5mm rostral to the obex in both the l-menthol (48.5+/-11.5 cells/section) and paraffin oil (26.0+/-9.6 cells/section) groups. Menthol 119-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 18343574-6 2008 In the SPV, the density of Fos-immunoreactive cells was highest at approximately 0.5mm rostral to the obex in both the l-menthol (48.5+/-11.5 cells/section) and paraffin oil (26.0+/-9.6 cells/section) groups. paraffin oils 161-173 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-30 18343574-7 2008 In the KF, the mean density of Fos-immunoreactive cells was highest at approximately 5.0mm rostral to the obex in both groups (l-menthol: 67.8+/-14.0 cells/section, control: 41.0+/-12.7 cells/section). Menthol 127-136 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 18349210-0 2008 Suppression of noxious-induced c-fos expression in the rat lumbar spinal cord by isoflurane alone or combined with fentanyl. Fentanyl 115-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-36 18349210-3 2008 Since the anesthetic mechanisms differ between inhaled anesthetics and opioids, we evaluated the differential effects of isoflurane and fentanyl on c-fos expression at the lumbar level as a measure of nociceptive information transfer during general anesthesia. Isoflurane 121-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 18349210-3 2008 Since the anesthetic mechanisms differ between inhaled anesthetics and opioids, we evaluated the differential effects of isoflurane and fentanyl on c-fos expression at the lumbar level as a measure of nociceptive information transfer during general anesthesia. Fentanyl 136-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 148-153 18349210-6 2008 RESULTS: The main suppressive effects on lumbar c-fos expression of isoflurane were observed in the superficial lamina II (P = 0.02), whereas fentanyl showed the strongest effects in lamina V (P = 0.05). Isoflurane 68-78 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-53 17941051-3 2008 The "promiscuous" IEGs Egr1 and c-fos, induced by growth factors and neurotrophins, in addition to depolarization, require activation of the MAP kinase signaling pathway for induction in response to KCl depolarization in PC12 cells; MEK1/2 inhibitors block KCl-induced Egr1 and c-fos expression. Potassium Chloride 199-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 17647273-2 2008 Here we showed that stress-inducing agent anisomycin was able to override the c-fos repression and to induce c-fos transcription in E1A + ras transformants. Anisomycin 42-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 17647273-2 2008 Here we showed that stress-inducing agent anisomycin was able to override the c-fos repression and to induce c-fos transcription in E1A + ras transformants. Anisomycin 42-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 109-114 17647273-3 2008 In vitro kinase assay data demonstrated that anisomycin increased phosphorylation of transactivation domain of Elk-1 transcription factor--a key regulator of inducible c-fos transcription. Anisomycin 45-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-173 17647273-5 2008 The activating effect of anisomycin on c-fos transcription could be abrogated by a prior treatment with N-acetyl-L-cysteine. Anisomycin 25-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 17647273-5 2008 The activating effect of anisomycin on c-fos transcription could be abrogated by a prior treatment with N-acetyl-L-cysteine. Acetylcysteine 104-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 39-44 17647273-7 2008 As anisomycin did not cause acetylation of nucleosome core histones, the present work focuses on the molecular mechanisms mediating the HDAC-independent induction of IEG c-fos by anisomycin in E1A + cHa-ras-transformed fibroblasts. Anisomycin 179-189 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 170-175 17941051-3 2008 The "promiscuous" IEGs Egr1 and c-fos, induced by growth factors and neurotrophins, in addition to depolarization, require activation of the MAP kinase signaling pathway for induction in response to KCl depolarization in PC12 cells; MEK1/2 inhibitors block KCl-induced Egr1 and c-fos expression. Potassium Chloride 199-202 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 278-283 17941051-3 2008 The "promiscuous" IEGs Egr1 and c-fos, induced by growth factors and neurotrophins, in addition to depolarization, require activation of the MAP kinase signaling pathway for induction in response to KCl depolarization in PC12 cells; MEK1/2 inhibitors block KCl-induced Egr1 and c-fos expression. Potassium Chloride 257-260 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 32-37 18164822-3 2008 Group ABA showed renewal of extinguished cocaine-seeking associated with c-Fos induction in basolateral amygdala, lateral hypothalamus, and infralimbic prefrontal cortex. alisol B 23-acetate 6-9 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 18164822-3 2008 Group ABA showed renewal of extinguished cocaine-seeking associated with c-Fos induction in basolateral amygdala, lateral hypothalamus, and infralimbic prefrontal cortex. Cocaine 41-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 18164822-4 2008 Groups ABA and ABB showed test-associated c-Fos induction in prelimbic prefrontal cortex, nucleus accumbens (core, shell, rostral pole), striatum, lateral amygdala, perifornical hypothalamus, and ventral tegmental area. alisol B 23-acetate 7-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 18164822-4 2008 Groups ABA and ABB showed test-associated c-Fos induction in prelimbic prefrontal cortex, nucleus accumbens (core, shell, rostral pole), striatum, lateral amygdala, perifornical hypothalamus, and ventral tegmental area. ABB 15-18 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 18314485-3 2008 In this study, we show that permanent Rat-1 cell lines genetically altered to overexpress c-Fos also displayed a similar MMRP to H(2)O(2), etoposide, and cisplatin as OC-14 cells. Cisplatin 154-163 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 17723286-0 2008 Effect of postnatal treadmill exercise on c-Fos expression in the hippocampus of rat pups born from the alcohol-intoxicated mothers. Alcohols 104-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 17723286-4 2008 In the present study, we investigated the influence of postnatal treadmill running on the c-Fos expression in the hippocampus of rat pups born from the alcohol-intoxicated mothers. Alcohols 152-159 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 18411708-2 2008 We demonstrated the differential patterns of c-fos mRNA induction by chemical somatic (formalin) and visceral (acetic acid) noxious stimuli in the rat amygdaloid nuclei, the brain regions implicated in emotion. Formaldehyde 87-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 18411708-2 2008 We demonstrated the differential patterns of c-fos mRNA induction by chemical somatic (formalin) and visceral (acetic acid) noxious stimuli in the rat amygdaloid nuclei, the brain regions implicated in emotion. Acetic Acid 111-122 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-50 18361829-0 2008 [Effects of teprenone on expression of heat shock protein 70 and c-fos in stomach following prednisolone ingestion: experiment with rats]. geranylgeranylacetone 12-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 18234123-12 2008 Expression of c-Fos was significantly decreased in the loxoprofen-Na group compared with the control group (p < 0.05). loxoprofen 55-65 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 18361829-1 2008 OBJECTIVE: To estimate the effect of teprenone on the expression of heat shock protein (HSP) 70 and c-fos in stomach following prednisolone ingestion. geranylgeranylacetone 37-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 18361829-1 2008 OBJECTIVE: To estimate the effect of teprenone on the expression of heat shock protein (HSP) 70 and c-fos in stomach following prednisolone ingestion. Prednisolone 127-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-105 18361829-10 2008 The expression value of c-fos of the model control group was 42 +/- 8, significantly higher than that of the normal control group (P < 0.01), and that expression values of c-fos of the 3 teprenone groups were all significantly lower than that of the model control groups dose-dependently (all P < 0.01). geranylgeranylacetone 190-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 18361829-10 2008 The expression value of c-fos of the model control group was 42 +/- 8, significantly higher than that of the normal control group (P < 0.01), and that expression values of c-fos of the 3 teprenone groups were all significantly lower than that of the model control groups dose-dependently (all P < 0.01). geranylgeranylacetone 190-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 175-180 18361829-12 2008 CONCLUSION: Teprenone is Pretreatment with teprenone, beneficial cytoprotective agent of gastric mucosa, has a gastroprotective effect against steroid-induced mucosal damage to a certain extent with the mechanism related to c-fos and HSP70 l level in the gastric mucosa. geranylgeranylacetone 12-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 224-229 18361829-12 2008 CONCLUSION: Teprenone is Pretreatment with teprenone, beneficial cytoprotective agent of gastric mucosa, has a gastroprotective effect against steroid-induced mucosal damage to a certain extent with the mechanism related to c-fos and HSP70 l level in the gastric mucosa. geranylgeranylacetone 43-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 224-229 18197100-11 2008 The direct compression of L5 nerve root produced an obvious expression of Fos-like immunoreactivity neurons in the dorsal horn of the spinal cord, which was significantly decreased by pretreatment with capsaicin. Capsaicin 202-211 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 18197387-5 2008 Increases in the quantity of c-Fos-positive cells 2 h after administration of lipopolysaccharide were seen in the following hypothalamic structures: AHN, PVH, LHA, VMH, DMH, and PH. Dimenhydrinate 169-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 18197387-6 2008 After electrical pain stimulation, the number of c-Fos-positive cells increased in these same hypothalamic structures (AHN, PVH, LHA, VMH, DMH, and PH). Dimenhydrinate 139-142 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 49-54 18005951-5 2008 Animal studies conducted on streptozotocin-induced diabetic rats suggested that the use of FOS as an alternative non-nutrient sweetener is without any adverse effects on various diabetes-related metabolic parameters. Streptozocin 28-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 91-94 17920665-8 2008 Additionally, we mapped Fos induction in rats treated with scopolamine and/or Ro 4368554; scopolamine increased Fos expression in the central nucleus of the amygdala and this was attenuated by Ro 4368554. Scopolamine 59-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 17920581-2 2008 Previous studies have shown that modafinil produces a different pattern of c-Fos activation in the brain to the classical stimulants amphetamine and methylphenidate. Modafinil 33-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 17920581-2 2008 Previous studies have shown that modafinil produces a different pattern of c-Fos activation in the brain to the classical stimulants amphetamine and methylphenidate. Amphetamine 133-144 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-80 18197392-0 2008 hnRNP-R regulates the PMA-induced c-fos expression in retinal cells. Tetradecanoylphorbol Acetate 22-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 34-39 18093170-6 2008 Although only 2% of striatal neurons were FosB labeled, 87% of these FosB-labeled neurons were co-labeled with c-fos when cocaine was injected in the cocaine-paired environment. Cocaine 122-129 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 111-116 18093170-8 2008 Furthermore, the total number of c-fos-labeled neurons was greater with either cocaine or saline challenge injections in the cocaine-paired environment than in the saline-paired environment. Cocaine 79-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 18093170-8 2008 Furthermore, the total number of c-fos-labeled neurons was greater with either cocaine or saline challenge injections in the cocaine-paired environment than in the saline-paired environment. Sodium Chloride 90-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 18093170-8 2008 Furthermore, the total number of c-fos-labeled neurons was greater with either cocaine or saline challenge injections in the cocaine-paired environment than in the saline-paired environment. Cocaine 125-132 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 18093170-8 2008 Furthermore, the total number of c-fos-labeled neurons was greater with either cocaine or saline challenge injections in the cocaine-paired environment than in the saline-paired environment. Sodium Chloride 164-170 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 33-38 18006871-7 2008 CIH led to significant cell injury in the left myocardium, including elevated LV myocyte size, measured by cell surface area (CIH 3,564 +/- 354 vs. HC 2,628 +/- 242 microm(2), P < 0.05) and cell length (CIH 148 +/- 23 vs. HC 115 +/- 16 microm, P < 0.05), elevated terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL)-stained positive cell number (CIH 98 +/- 45 vs. HC 15 +/- 13, P < 0.01), elevated caspase-3 activity (906 +/- 249 vs. 2,275 +/- 1,169 pmol x min(-1) x mg(-1), P < 0.05), and elevated expression of several remodeling gene markers, including c-fos, atrial natriuretic peptide, beta-myosin heavy chain, and myosin light chain-2. cih 0-3 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 594-599 17673334-7 2008 A low tryptophan diet led to decreases in plasma tryptophan levels, low ratio of tryptophan/large neutral amino acid, whole blood 5-HT, and neuronal 5-HT content in the Dorsal and Median Raphe Nuclei, as well as altered c-fos expression in the brain. Tryptophan 6-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 220-225 17934249-2 2008 Since Fos immunolabeling of the SPO of rats treated by caudal fourth ventricular (CV4) administration of the glucose antimetabolite, 5-thioglucose (5TG), parallels the distribution of GABA neuronal perikarya, the current studies investigated the genomic responsiveness of neuroanatomically-defined populations of glutamate decarboxylase (GAD)-immunoreactive (-ir) neurons in this region of the brain to hindbrain glucoprivation. Glucose 109-116 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 6-9 17934249-2 2008 Since Fos immunolabeling of the SPO of rats treated by caudal fourth ventricular (CV4) administration of the glucose antimetabolite, 5-thioglucose (5TG), parallels the distribution of GABA neuronal perikarya, the current studies investigated the genomic responsiveness of neuroanatomically-defined populations of glutamate decarboxylase (GAD)-immunoreactive (-ir) neurons in this region of the brain to hindbrain glucoprivation. 5-thio-D-glucose 133-146 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 6-9 17934249-6 2008 SPO GABA neurons in the vehicle-treated controls were uniformly Fos-ir-negative. gamma-Aminobutyric Acid 4-8 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 18281894-1 2008 We studied Fos immunoreactivity within targets of the mesocorticolimbic dopamine systems at different stages of acquisition of heroin place conditioning. Dopamine 72-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 18281894-4 2008 Further, the medial prefrontal cortex was the only region displaying higher Fos expression in the group exposed to both heroin and conditioning compartment. Heroin 120-126 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 76-79 17963758-1 2008 Marked hippocampal changes in response to excitatory amino acid agonists occur during pregnancy (e.g. decreased frequency in spontaneous recurrent seizures in rats with KA lesions of the hippocampus) and lactation (e.g. reduced c-Fos expression in response to N-methyl-d,l-aspartic acid but not to kainic acid). Excitatory Amino Acids 42-63 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 228-233 17920665-8 2008 Additionally, we mapped Fos induction in rats treated with scopolamine and/or Ro 4368554; scopolamine increased Fos expression in the central nucleus of the amygdala and this was attenuated by Ro 4368554. Scopolamine 59-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 17920665-8 2008 Additionally, we mapped Fos induction in rats treated with scopolamine and/or Ro 4368554; scopolamine increased Fos expression in the central nucleus of the amygdala and this was attenuated by Ro 4368554. Scopolamine 90-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-27 17920665-8 2008 Additionally, we mapped Fos induction in rats treated with scopolamine and/or Ro 4368554; scopolamine increased Fos expression in the central nucleus of the amygdala and this was attenuated by Ro 4368554. Scopolamine 90-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 112-115 17884736-4 2007 The total number of c-Fos immunopositive cells was counted in the dorsal motor nucleus of the vagus (DMV), the nucleus tractus solitarius (NTS) and the rostral ventrolateral medulla (RVLM). (2R,3R)-2,3-dihydroxy-3-methylpentanoic acid 101-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 18390209-0 2008 [Effects of preemptively injected intrathecal lornoxicam on behavior and c-fos protein expression of rat with formalin hurting]. Formaldehyde 110-118 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 73-78 18390209-1 2008 OBJECTIVE: The objective of this study was to investigate an antinociceptive effect of preemptive intrathecal lornoxicam on behavior and expression of c-Fos protein in the spinal cord of the formalin hurting rats. lornoxicam 110-120 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-156 18390209-1 2008 OBJECTIVE: The objective of this study was to investigate an antinociceptive effect of preemptive intrathecal lornoxicam on behavior and expression of c-Fos protein in the spinal cord of the formalin hurting rats. Formaldehyde 191-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 151-156 18390209-16 2008 CONCLUSION: Pre-intrathacal lornoxicam can successfully inhibit nociceptive behavior and c-Fos expression in spinal dorsal horn of formalin test rats. lornoxicam 28-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 18390209-16 2008 CONCLUSION: Pre-intrathacal lornoxicam can successfully inhibit nociceptive behavior and c-Fos expression in spinal dorsal horn of formalin test rats. Formaldehyde 131-139 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-94 18589755-0 2008 [Effects of beta-asarone on expression of FOS and GAD65 in cortex of epileptic rat induced by penicillin]. asarone 12-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 18159243-5 2007 We confirm, in a series of experiments comprised of cFos staining, behavioral analysis and neurochemical determinations, that ketamine interferes with the behavioral expression of fear and with normal fear processing in the amygdala and related brain regions. Ketamine 126-134 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-56 17826765-6 2007 Fos-immunoreactivity induced by acute apomorphine administration (barrel cortex, paraventricular hypothalamic nucleus, central amygdala and locus coeruleus) was strongly reduced after chronic administration. Apomorphine 38-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 17826765-8 2007 Apomorphine, however, reduced mating-induced Fos-immunoreactivity in the nucleus accumbens shell and prevented its occurrence in its core area. Apomorphine 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 17823256-7 2007 The effect of estradiol on intraduodenal Intralipid-induced satiation was mirrored by selective increases in the number of cells expressing c-Fos immunoreactivity in a circumscribed region of the nucleus tractus solitarius (NTS), just caudal to the area postrema (cNTS) but not elsewhere in the NTS or the hypothalamic paraventricular or arcuate nuclei. Estradiol 14-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 17823256-7 2007 The effect of estradiol on intraduodenal Intralipid-induced satiation was mirrored by selective increases in the number of cells expressing c-Fos immunoreactivity in a circumscribed region of the nucleus tractus solitarius (NTS), just caudal to the area postrema (cNTS) but not elsewhere in the NTS or the hypothalamic paraventricular or arcuate nuclei. soybean oil, phospholipid emulsion 41-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-145 17720257-0 2007 Repeated amphetamine administration outside the home cage enhances drug-induced Fos expression in rat nucleus accumbens. Amphetamine 9-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 17720257-1 2007 Induction of the immediate early gene protein product Fos has been used extensively to assess neural activation in the striatum after repeated amphetamine administration to rats in their home cages. Amphetamine 143-154 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 54-57 17720257-3 2007 We determined the dose-response relationship for amphetamine-induced psychomotor activity and Fos expression in nucleus accumbens and caudate-putamen 1 week after repeated administration of amphetamine or saline in locomotor activity chambers. Amphetamine 49-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 17720257-3 2007 We determined the dose-response relationship for amphetamine-induced psychomotor activity and Fos expression in nucleus accumbens and caudate-putamen 1 week after repeated administration of amphetamine or saline in locomotor activity chambers. Amphetamine 190-201 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-97 17720257-4 2007 Repeated administration of amphetamine enhanced amphetamine-induced locomotor activity and stereotypy and Fos expression in nucleus accumbens, but not in caudate-putamen. Amphetamine 27-38 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 17720257-5 2007 In comparison, levels of Fos expression induced by 1mg/kg amphetamine were not altered in nucleus accumbens or caudate-putamen by repeated amphetamine administration in the home cage. Amphetamine 58-69 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-28 17720257-7 2007 Furthermore, repeated amphetamine administration increased drug-induced Fos expression in enkephalin-positive, but not enkephalin-negative, neurons in nucleus accumbens. Amphetamine 22-33 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 18052972-6 2007 On the other hand, pair-exposure to the CS with an unfamiliar rat eliminated the behavioral, but not the autonomic, response, with Fos expression in the basal nucleus of the amygdala and infralimbic region of the prefrontal cortex. Cesium 40-42 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-134 18589755-0 2008 [Effects of beta-asarone on expression of FOS and GAD65 in cortex of epileptic rat induced by penicillin]. Penicillins 94-104 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 18589755-1 2008 OBJECTIVE: To study effects of beta-asarone on expression of FOS and GAD65 in cortex of epileptic rat induced by penicillin. asarone 31-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 18589755-1 2008 OBJECTIVE: To study effects of beta-asarone on expression of FOS and GAD65 in cortex of epileptic rat induced by penicillin. Penicillins 113-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-64 18589755-5 2008 The effects of beta-asarone on expression of FOS and GAD65 in cortex of epileptic rat were detected by immuohistochemistry method. asarone 15-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 45-48 18589755-6 2008 RESULTS: beta-asarone could raise expression of FOS and reduce expression of GAD65 obviously. asarone 9-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 48-51 18589755-9 2008 CONCLUSION: Up-regulation of FOS may be a effective link of anti-epileptic effect of beta-asarone; reduced expression of GAD65 may be a follow-up impact of beta-asarone treatment. asarone 85-97 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-32 18053328-1 2007 Repeated injections of nicotine can produce an increase in locomotor activity and the expression of immediate-early gene, c-fos, in the central dopaminergic areas. Nicotine 23-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 122-127 18053328-8 2007 significantly inhibited the nicotine-induced locomotor activity and expression of c-Fos in the striatum and the nucleus accumbens. Nicotine 28-36 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 17559977-6 2007 Western blot analysis revealed differential expression profiles of Fos family members in neurons briefly exposed to DCG-IV and NMDA. dcg 116-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 17559977-6 2007 Western blot analysis revealed differential expression profiles of Fos family members in neurons briefly exposed to DCG-IV and NMDA. N-Methylaspartate 127-131 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-70 17686536-0 2007 Olfactory discrimination ability and brain expression of c-fos, Gir and Glut1 mRNA are altered in n-3 fatty acid-depleted rats. Fatty Acids, Omega-3 98-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-62 18001278-0 2007 Effects of opioid receptor blockade on the renewal of alcohol seeking induced by context: relationship to c-fos mRNA expression. Alcohols 54-61 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-111 18001278-8 2007 Re-exposure to the alcohol-associated context (ABA) significantly increased operant behaviour on the previously active lever relative to the AAA groups and this increased responding was associated with increased c-fos mRNA expression in the basal and lateral amygdala and the CA3 subregion of the hippocampus. Alcohols 19-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 212-217 18001278-8 2007 Re-exposure to the alcohol-associated context (ABA) significantly increased operant behaviour on the previously active lever relative to the AAA groups and this increased responding was associated with increased c-fos mRNA expression in the basal and lateral amygdala and the CA3 subregion of the hippocampus. alisol B 23-acetate 47-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 212-217 18001278-9 2007 Naltrexone pre-treatment attenuated context-induced alcohol seeking and inhibited c-fos mRNA expression in the lateral amygdala and CA3. Naltrexone 0-10 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 82-87 17900567-3 2007 The present paper examines the Fos-protein distribution in the brain of rats submitted to a conditioned place aversion (CPA) paradigm using the dPAG chemical stimulation with semicarbazide (SMC), an inhibitor of the GABA synthesizing enzyme, as US and the quadrant of an arena where the drug was injected as the paired neutral stimulus. cpa 120-123 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 17900567-3 2007 The present paper examines the Fos-protein distribution in the brain of rats submitted to a conditioned place aversion (CPA) paradigm using the dPAG chemical stimulation with semicarbazide (SMC), an inhibitor of the GABA synthesizing enzyme, as US and the quadrant of an arena where the drug was injected as the paired neutral stimulus. dpag 144-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 17900567-3 2007 The present paper examines the Fos-protein distribution in the brain of rats submitted to a conditioned place aversion (CPA) paradigm using the dPAG chemical stimulation with semicarbazide (SMC), an inhibitor of the GABA synthesizing enzyme, as US and the quadrant of an arena where the drug was injected as the paired neutral stimulus. carbamylhydrazine 175-188 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 17900567-3 2007 The present paper examines the Fos-protein distribution in the brain of rats submitted to a conditioned place aversion (CPA) paradigm using the dPAG chemical stimulation with semicarbazide (SMC), an inhibitor of the GABA synthesizing enzyme, as US and the quadrant of an arena where the drug was injected as the paired neutral stimulus. carbamylhydrazine 190-193 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 17900567-3 2007 The present paper examines the Fos-protein distribution in the brain of rats submitted to a conditioned place aversion (CPA) paradigm using the dPAG chemical stimulation with semicarbazide (SMC), an inhibitor of the GABA synthesizing enzyme, as US and the quadrant of an arena where the drug was injected as the paired neutral stimulus. gamma-Aminobutyric Acid 216-220 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 31-34 17900567-4 2007 Our results show that CPA associated with SMC injections caused a significant Fos labeling in the laterodorsal nucleus of the thalamus (LD), basolateral nucleus of amygdala (BLA) and in the dorsomedial PAG (dmPAG). cpa 22-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 17900567-4 2007 Our results show that CPA associated with SMC injections caused a significant Fos labeling in the laterodorsal nucleus of the thalamus (LD), basolateral nucleus of amygdala (BLA) and in the dorsomedial PAG (dmPAG). carbamylhydrazine 42-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 17346888-4 2007 administration of NaHS, a H(2)S donor, produced prompt hyperalgesia in rats, accompanied by expression of Fos in the spinal dorsal horn. sodium bisulfide 18-22 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 17346888-4 2007 administration of NaHS, a H(2)S donor, produced prompt hyperalgesia in rats, accompanied by expression of Fos in the spinal dorsal horn. Hydrogen Sulfide 26-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 106-109 17853060-5 2007 Fos-AVP double labeling in the parvocellular medial paraventricular nucleus (PaMP) in ADX groups was increased after 3 h in basal conditions, and in all periods after restraint stress. pamp 77-81 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 17853060-7 2007 Fos expression was increased in the PaMP after 3 h and after restraint stress in the Sham and ADX groups, especially in the ADX group. pamp 36-40 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 17853060-9 2007 The Fos-AVP double labeling findings in the PaMP also indicate a minor participation of these vasopressinergic neurons in the regulation of the HPA axis after ADX. pamp 44-48 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 4-7 17920048-3 2007 A single cocaine administration (30 mg/kg) increased ERK-mediated signaling proteins, phosphoryation of cAMP response element-binding protein (CREB) kinase, pp90 ribosomal S6 kinase (RSK), and c-Fos protein levels in the caudate/putamen of Fischer rats. Cocaine 9-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 193-198 17884026-7 2007 The hyperalgesic response to the formalin test was accompanied by reduced Fos expression in the paraventricular nucleus of the hypothalamus, which is part of a network (the medial pain system) that mediates motivational-affective aspects of pain. Formaldehyde 33-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 74-77 17913375-0 2007 L-type calcium channel blockade on haloperidol-induced c-Fos expression in the striatum. Haloperidol 35-46 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 17913375-1 2007 Haloperidol-induced c-Fos expression in the lateral part of the neostriatum has been correlated with motor side effects while c-Fos induction in the medial part of the neostriatum and the nucleus accumbens is thought to be associated with the therapeutic effects of the drug. Haloperidol 0-11 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 20-25 17913375-2 2007 Induction of c-Fos in the striatum by haloperidol involves dopamine D(2) (DA D(2)) receptor antagonism and is dependent on activation of N-methyl-d-aspartate (NMDA) receptors and L-type Ca(2+) channels. Haloperidol 38-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 17913375-2 2007 Induction of c-Fos in the striatum by haloperidol involves dopamine D(2) (DA D(2)) receptor antagonism and is dependent on activation of N-methyl-d-aspartate (NMDA) receptors and L-type Ca(2+) channels. Dopamine 59-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 13-18 17913375-3 2007 In the current study, pretreatment with L-type Ca(2+) channel blockers suppressed haloperidol-induced c-Fos throughout the neostriatum and the nucleus accumbens at 2 h postinjection. Haloperidol 82-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-107 17913375-4 2007 However, elevated c-Fos protein expression was observed only in the lateral part of the neostriatum at 5 h postinjection of haloperidol following pretreatment of L-type Ca(2+) channel blocker compared with rats pretreated with vehicle alone. Haloperidol 124-135 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-23 17913375-6 2007 Infusions of L-type Ca(2+) channel blockers directly into the neostriatum mimicked similar patterns of changes in haloperidol-induced c-Fos expression. Haloperidol 114-125 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 134-139 17913375-7 2007 Prolonged expression of c-Fos was not observed following coadministration of nifedipine and a dopamine D(1) (DA D(1)) receptor agonist, SKF 81297, but could be mimicked by the DA D(2/3) receptor antagonist raclopride, suggesting that the phenomenon is likely related to DA D(2) receptor antagonism. Nifedipine 77-87 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 17913375-7 2007 Prolonged expression of c-Fos was not observed following coadministration of nifedipine and a dopamine D(1) (DA D(1)) receptor agonist, SKF 81297, but could be mimicked by the DA D(2/3) receptor antagonist raclopride, suggesting that the phenomenon is likely related to DA D(2) receptor antagonism. Raclopride 206-216 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 17931790-3 2007 The objective of this study was to characterize the effects of different amphetamine paradigms on the Fos activation of GABAergic interneurons that contain parvalbumin in the medial prefrontal cortex. Amphetamine 73-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 102-105 17931790-4 2007 Although a sensitizing, repeated regimen of amphetamine induced Fos in all cortical layers, only layer V parvalbumin-immunolabeled cells were activated in the infralimbic and prelimbic cortices. Amphetamine 44-55 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 17931790-6 2007 An acute amphetamine injection to naive rats was associated with an increase in Fos, but in parvalbumin-positive neurons of the prelimbic cortex, where it was preferentially induced in layer III. Amphetamine 9-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 80-83 17506118-0 2007 C-fos expression at the spinal dorsal horn of streptozotocin-induced diabetic rats. Streptozocin 46-60 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 17506118-2 2007 We used the activation of the c-fos protooncogene to study the activity of spinal dorsal horn neurons in streptozotocin (STZ)-induced diabetic rats in the absence of stimulation or in response to innocuous or noxious stimuli. Streptozocin 105-119 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 30-35 17506118-5 2007 RESULTS: In the absence of stimulation, STZ-injected rats presented significantly higher numbers of Fos-IR neurons than controls, both in the superficial and deep dorsal horn (DDH). Streptozocin 40-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 100-103 17506118-7 2007 Noxious mechanical and noxious thermal stimuli increased the numbers of Fos-IR neurons, both in control and STZ-rats, but in a more pronounced manner when diabetic rats were subjected to noxious mechanical stimulation. Streptozocin 108-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 17970739-1 2007 We tested the hypothesis that amphetamine (AMPH)-induced conditioned motor sensitization is accompanied by cellular activation (measured by Fos immunoreactivity) and synaptophysin immunoreactivity in reward-related brain areas. Amphetamine 30-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-143 17970739-1 2007 We tested the hypothesis that amphetamine (AMPH)-induced conditioned motor sensitization is accompanied by cellular activation (measured by Fos immunoreactivity) and synaptophysin immunoreactivity in reward-related brain areas. Amphetamine 43-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 140-143 17970739-6 2007 AMPH administered in the AMPH-paired context increased the density of both Fos and synaptophysin immunoreactivity in the dentate gyrus, cornu ammonis (CA)1, CA3, basolateral amygdala and dorsolateral striatum. Amphetamine 0-4 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 75-78 17970739-8 2007 Saline administered in the AMPH-paired context increased the density of Fos immunoreactivity in the basolateral amygdala and nucleus accumbens core. Sodium Chloride 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 72-75 17299504-6 2007 Cocaine-induced c-fos mRNA was similar across ages in the dorsal caudate putamen (CPu), nucleus accumbens, and lateral bed nucleus of the stria terminalis. Cocaine 0-7 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 16-21 17698179-0 2007 Subchronic phencyclidine exposure potentiates the behavioral and c-Fos response to stressful stimuli in rats. Phencyclidine 11-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 65-70 17928140-6 2007 Injection of 5% formalin into the MF at levels L4 and L5 induced a significant increase in the number of c-Fos-immunoreactive (-ir) nuclei in the dorsal horn in many lumbar segments. Formaldehyde 16-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 17928140-10 2007 The number of double-labeled dorsal horn neurons (FG-positive plus c-Fos-ir) in all lumbar segments was significantly higher in the formalin group than in the saline group. Formaldehyde 132-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 67-72 17628002-6 2007 In contrast, the effect of TSA on the norepinephrine induction of the c-fos mRNA was stimulatory. trichostatin A 27-30 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 17670858-6 2007 A terminal c-Fos study revealed intact LiCl-induced c-Fos expression in the lateral parabrachial nucleus and central amygdala in DSAP rats, despite significant loss of NST NA neurons and attenuated c-Fos activation of corticotropin-releasing hormone-positive neurons in the paraventricular nucleus of the hypothalamus (PVN). Lithium Chloride 39-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 17670858-6 2007 A terminal c-Fos study revealed intact LiCl-induced c-Fos expression in the lateral parabrachial nucleus and central amygdala in DSAP rats, despite significant loss of NST NA neurons and attenuated c-Fos activation of corticotropin-releasing hormone-positive neurons in the paraventricular nucleus of the hypothalamus (PVN). Lithium Chloride 39-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 17670858-6 2007 A terminal c-Fos study revealed intact LiCl-induced c-Fos expression in the lateral parabrachial nucleus and central amygdala in DSAP rats, despite significant loss of NST NA neurons and attenuated c-Fos activation of corticotropin-releasing hormone-positive neurons in the paraventricular nucleus of the hypothalamus (PVN). Lithium Chloride 39-43 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 17881916-1 2007 PURPOSE: To examine the expression pattern of the stress-related genes c-fos and c-jun, which encode the 2 major components of activator protein (AP)-1, and cyclooxygenase (COX)-2 in rat corneal epithelium treated with topical antiglaucoma medications and benzalkonium chloride (BAK) preservative. Benzalkonium Compounds 256-277 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 17881916-1 2007 PURPOSE: To examine the expression pattern of the stress-related genes c-fos and c-jun, which encode the 2 major components of activator protein (AP)-1, and cyclooxygenase (COX)-2 in rat corneal epithelium treated with topical antiglaucoma medications and benzalkonium chloride (BAK) preservative. Benzalkonium Compounds 279-282 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 17881916-10 2007 Expression of c-fos and c-jun seemed more marked with prostaglandins than with timolol. Prostaglandins 54-68 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 17881916-10 2007 Expression of c-fos and c-jun seemed more marked with prostaglandins than with timolol. Timolol 79-86 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 17881916-11 2007 Thirty minutes to 1 hour after instillation of 0.02% BAK preservative, signals for c-fos and c-jun mRNA were detected in the corneal and conjunctival epithelium. Benzalkonium Compounds 53-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 17466093-9 2007 Moreover, dietary taurine inhibited activator protein-1 (AP-1) (c-fos/c-jun subunits) expression (P < 0.05), up regulated NF-kappaB (p65) expression (P < 0.05), and decreased caspase-1 expression (P < 0.05) so as to reduce the apoptosis of photoreceptors in the retina (P < 0.05). Taurine 18-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-69 17628002-6 2007 In contrast, the effect of TSA on the norepinephrine induction of the c-fos mRNA was stimulatory. Norepinephrine 38-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-75 17628002-8 2007 Chromatin immunoprecipitation with antibodies against acetylated Lys14 of H3 showed an increase in DNA recovery of the promoter regions of aa-nat, mkp-1, and c-fos after treatment with TSA. trichostatin A 185-188 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 158-163 17927775-5 2007 However, retrogradely labeled neurons positive for c-fos were increased only in the dorsal DMH and adjoining region in both stressed and lipopolysaccharide-treated groups, and triple-labeled neurons were found only in this area in rats subjected to either stress paradigm. 1,2-Dimethylhydrazine 91-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 51-56 17883415-7 2007 In another experiment, lactating rats were treated as above, except that 90 min after the end of the observation period the rats were killed and their brains were processed for immunohistochemical detection of Fos protein in the PAG. pag 229-232 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 210-213 17927775-6 2007 Thus, hypothalamic neurons that project to the rRP and express c-fos in response to either experimental stress or systemic inflammation are found only in the dorsal DMH, and many of those activated by stress contain nNOS, suggesting that nitric oxide may play a role in signaling in this pathway. 1,2-Dimethylhydrazine 165-168 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 17927775-6 2007 Thus, hypothalamic neurons that project to the rRP and express c-fos in response to either experimental stress or systemic inflammation are found only in the dorsal DMH, and many of those activated by stress contain nNOS, suggesting that nitric oxide may play a role in signaling in this pathway. Nitric Oxide 238-250 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 63-68 17663450-3 2007 After 8-day dietary sodium deprivation, immunoreactivity for c-Fos (a neuronal activity marker) increased markedly within the aldosterone-sensitive neurons of the NTS, which express the enzyme 11-beta-hydroxysteroid dehydrogenase type 2 (HSD2). Sodium 20-26 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 17663450-5 2007 Then, 1-2 hours after sodium-deprived rats ingested salt (a hypertonic 3% solution of NaCl), c-Fos immunoreactivity within the HSD2 neurons was virtually eliminated, despite a large increase in c-Fos activation in the surrounding NTS (including the A2 noradrenergic neurons) and area postrema. Salts 52-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 17663450-5 2007 Then, 1-2 hours after sodium-deprived rats ingested salt (a hypertonic 3% solution of NaCl), c-Fos immunoreactivity within the HSD2 neurons was virtually eliminated, despite a large increase in c-Fos activation in the surrounding NTS (including the A2 noradrenergic neurons) and area postrema. Salts 52-56 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 194-199 17663450-5 2007 Then, 1-2 hours after sodium-deprived rats ingested salt (a hypertonic 3% solution of NaCl), c-Fos immunoreactivity within the HSD2 neurons was virtually eliminated, despite a large increase in c-Fos activation in the surrounding NTS (including the A2 noradrenergic neurons) and area postrema. Sodium Chloride 86-90 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 93-98 17663450-6 2007 Also after salt intake, c-Fos activation increased within pontine nuclei that relay gustatory (caudal medial PB) and viscerosensory (rostral lateral PB) information from the NTS to the forebrain. Salts 11-15 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 24-29 17850462-9 2007 Lard-associated changes in c-Fos(+) cell numbers were observed in the nucleus of the tractus solitarius, lateral parabrachial nucleus, central nucleus of the amygdala, ventral tegmental area, nucleus accumbens shell and the prefrontal cortex, and were associated with lower levels of triglycerides and free fatty acids. Triglycerides 284-297 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 17850462-9 2007 Lard-associated changes in c-Fos(+) cell numbers were observed in the nucleus of the tractus solitarius, lateral parabrachial nucleus, central nucleus of the amygdala, ventral tegmental area, nucleus accumbens shell and the prefrontal cortex, and were associated with lower levels of triglycerides and free fatty acids. Fatty Acids, Nonesterified 302-318 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 17937860-0 2007 [Effect of carbenoxolone on expression of Fos, NMDAR2 and GFAP in the hippocampus of pentylenetetrazo-kindled epileptic rats]. Carbenoxolone 11-24 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 17937860-0 2007 [Effect of carbenoxolone on expression of Fos, NMDAR2 and GFAP in the hippocampus of pentylenetetrazo-kindled epileptic rats]. pentylenetetrazo 85-101 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-45 17937860-10 2007 The NMDAR2-Li and Fos-Li neurons as well as GFAP-Li astrocytes in hippocampi increased in PTZ-kindled epileptic rats compared with controls. Pentylenetetrazole 90-93 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 18-21 17937860-13 2007 CONCLUSIONS: CBX may inhibit spontaneous seizures and decrease the numbers of Fos-Li and NMDARZ-Li neurons, thus providing anti-epileptic effects. Carbenoxolone 13-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-81 17822683-2 2007 The present experiment explored the nature of this plasticity by quantifying Fos immunoreactivity (Fos-ir) in structures implicated in the mediation of sodium appetite and in the signaling of reward. Sodium 152-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-80 17822683-2 2007 The present experiment explored the nature of this plasticity by quantifying Fos immunoreactivity (Fos-ir) in structures implicated in the mediation of sodium appetite and in the signaling of reward. Sodium 152-158 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-102 17628530-7 2007 The inhibitory effect of retigabine on psychostimulant-induced locomotor activity was accompanied by a marked reduction in c-Fos expression, in particular the nucleus accumbens and primary motor cortex were responsive to retigabine pre-treatment. ezogabine 25-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 123-128 17785415-8 2007 PCP increased c-fos expression in PFC pyramidal neurons, an effect prevented by the administration of clozapine. Clozapine 102-111 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 14-19 17693029-2 2007 Here we examined the activation of Fos immunoreactivity (Fos-IR) following exposure to a CS previously paired with either paced or nonpaced copulation. Cesium 89-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-38 17693029-2 2007 Here we examined the activation of Fos immunoreactivity (Fos-IR) following exposure to a CS previously paired with either paced or nonpaced copulation. Cesium 89-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 57-60 17693029-9 2007 In both experiments, the CS associated with paced copulation produced significantly more Fos-IR in the piriform cortex, medial preoptic area, and ventral tegmental area, relative to the same odor or strain cues associated with nonpaced copulation. Cesium 25-27 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 89-92 17567719-5 2007 Sodium intake induced by PD significantly increased the number of Fos-5-HT cells, independently of the concentration of NaCl consumed. Sodium 0-6 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 66-69 17827730-9 2007 Cp-Mn and DiAc-Cp-Mn treatment significantly decreased c-Fos expression elicited by KA. cp-mn 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 17827730-9 2007 Cp-Mn and DiAc-Cp-Mn treatment significantly decreased c-Fos expression elicited by KA. diac-cp-mn 10-20 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 55-60 17663450-3 2007 After 8-day dietary sodium deprivation, immunoreactivity for c-Fos (a neuronal activity marker) increased markedly within the aldosterone-sensitive neurons of the NTS, which express the enzyme 11-beta-hydroxysteroid dehydrogenase type 2 (HSD2). Aldosterone 126-137 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 61-66 17240043-9 2007 Finally, cyclophosphamide increased c-fos expression in the rat lumbar spinal cord. Cyclophosphamide 9-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 36-41 17594906-0 2007 The effects of acute corticosterone administration on anxiety, endogenous corticosterone, and c-Fos expression in the rat brain. Corticosterone 21-35 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 94-99 17762649-5 2007 The PKC inhibitor antagonized angiotensin II-induced p47 phosphorylation, and an antisense oligonucleotide (ASON) complementary to PKCbeta messenger RNA suppressed angiotensin II-induced ERK1/2 activation, phosphorylation or DNA binding activity of CREB, and upregulation of c-fos mRNA expression in the ventrolateral medulla. Oligonucleotides 91-106 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 275-280 17762649-5 2007 The PKC inhibitor antagonized angiotensin II-induced p47 phosphorylation, and an antisense oligonucleotide (ASON) complementary to PKCbeta messenger RNA suppressed angiotensin II-induced ERK1/2 activation, phosphorylation or DNA binding activity of CREB, and upregulation of c-fos mRNA expression in the ventrolateral medulla. Oligonucleotides, Antisense 108-112 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 275-280 17881193-6 2007 c-Fos protein was expressed in most neurons with minimum content of orexin, i.e. activation of these neurons correlated with the redistribution of orexins caused by skin EHF-irradiation and injection of cyclophosphamide (CPA). Cyclophosphamide 203-219 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5 17881193-6 2007 c-Fos protein was expressed in most neurons with minimum content of orexin, i.e. activation of these neurons correlated with the redistribution of orexins caused by skin EHF-irradiation and injection of cyclophosphamide (CPA). Cyclophosphamide 221-224 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-5