PMID-sentid Pub_year Sent_text comp_official_name comp_offset protein_name organism prot_offset 20861370-3 2010 Orexin B (1 muM) induced depolarization in orexin neurons, which was still observed in the presence of TTX (1 muM), AP-5 (50 muM), and CNQX (20 muM). Tetrodotoxin 103-106 hypocretin Mus musculus 0-8 20861370-3 2010 Orexin B (1 muM) induced depolarization in orexin neurons, which was still observed in the presence of TTX (1 muM), AP-5 (50 muM), and CNQX (20 muM). Tetrodotoxin 103-106 hypocretin Mus musculus 43-49 20861370-3 2010 Orexin B (1 muM) induced depolarization in orexin neurons, which was still observed in the presence of TTX (1 muM), AP-5 (50 muM), and CNQX (20 muM). 2-amino-5-phosphopentanoic acid 116-120 hypocretin Mus musculus 0-8 20861370-3 2010 Orexin B (1 muM) induced depolarization in orexin neurons, which was still observed in the presence of TTX (1 muM), AP-5 (50 muM), and CNQX (20 muM). 2-amino-5-phosphopentanoic acid 116-120 hypocretin Mus musculus 43-49 20861370-3 2010 Orexin B (1 muM) induced depolarization in orexin neurons, which was still observed in the presence of TTX (1 muM), AP-5 (50 muM), and CNQX (20 muM). 6-Cyano-7-nitroquinoxaline-2,3-dione 135-139 hypocretin Mus musculus 0-8 20861370-3 2010 Orexin B (1 muM) induced depolarization in orexin neurons, which was still observed in the presence of TTX (1 muM), AP-5 (50 muM), and CNQX (20 muM). 6-Cyano-7-nitroquinoxaline-2,3-dione 135-139 hypocretin Mus musculus 43-49 20861370-4 2010 In addition, orexin B induced inward currents in the presence of TTX, suggesting a direct activation of orexin neurons. Tetrodotoxin 65-68 hypocretin Mus musculus 13-21 20861370-4 2010 In addition, orexin B induced inward currents in the presence of TTX, suggesting a direct activation of orexin neurons. Tetrodotoxin 65-68 hypocretin Mus musculus 13-19 19915095-0 2010 The modulation of orexin A on HCN currents of pyramidal neurons in mouse prelimbic cortex. Hydrogen Cyanide 30-33 hypocretin Mus musculus 18-26 20547681-2 2010 Hypothalamic orexin neurons are CO(2) sensitive in vitro, prepro-orexin knockout mice have a reduced CO(2) response prominently in wakefulness, and focal antagonism of the orexin receptor 1 (OX(1)R) in two central chemoreceptor sites, the retrotrapezoid nucleus (RTN) or the medullary raphe, results in a reduction of the CO(2) response predominately in wakefulness (-30% and -16%, respectively). co(2) 32-37 hypocretin Mus musculus 13-19 20547681-2 2010 Hypothalamic orexin neurons are CO(2) sensitive in vitro, prepro-orexin knockout mice have a reduced CO(2) response prominently in wakefulness, and focal antagonism of the orexin receptor 1 (OX(1)R) in two central chemoreceptor sites, the retrotrapezoid nucleus (RTN) or the medullary raphe, results in a reduction of the CO(2) response predominately in wakefulness (-30% and -16%, respectively). co(2) 101-106 hypocretin Mus musculus 58-71 20547681-2 2010 Hypothalamic orexin neurons are CO(2) sensitive in vitro, prepro-orexin knockout mice have a reduced CO(2) response prominently in wakefulness, and focal antagonism of the orexin receptor 1 (OX(1)R) in two central chemoreceptor sites, the retrotrapezoid nucleus (RTN) or the medullary raphe, results in a reduction of the CO(2) response predominately in wakefulness (-30% and -16%, respectively). co(2) 101-106 hypocretin Mus musculus 58-71 20547681-3 2010 Here we hypothesize that acute and selective inhibition of both orexin receptors (OX(1)R and OX(2)R) at all central locations by an orally administered dual orexin receptor antagonist, almorexant, will substantially attenuate the CO(2) response in a vigilance-state- and diurnal-cycle-dependent manner. co(2) 230-235 hypocretin Mus musculus 64-70 19915095-6 2010 The excitatory effect of orexin A on PL pyramidal neurons was enhanced when HCN currents were diminished, while attenuated when HCN currents were enlarged. Hydrogen Cyanide 128-131 hypocretin Mus musculus 25-33 19915095-3 2010 In the present study, using whole-cell voltage clamp recordings, the effect of an arousal peptide, orexin A, on HCN currents in layer V pyramidal neurons from mouse prelimbic cortex (PL), the homolog of the prefrontal cortex was investigated. Hydrogen Cyanide 112-115 hypocretin Mus musculus 99-107 19915095-7 2010 In summary, orexin A inhibits HCN currents and enhances excitability of pyramidal neurons in PL, which may contribute to arousal and cognition. Hydrogen Cyanide 30-33 hypocretin Mus musculus 12-20 19915095-4 2010 The results demonstrated that orexin A suppressed HCN currents and shifted their activation curve to a more negative direction. Hydrogen Cyanide 50-53 hypocretin Mus musculus 30-38 19915095-5 2010 This action of orexin A was blocked by SB334867, an orexin receptor 1 (OXR1) blocker and bisindolylmaleimide, a protein kinase C (PKC) inhibitor, indicating the involvement of OXR1 and PKC. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 39-47 hypocretin Mus musculus 15-23 19915095-5 2010 This action of orexin A was blocked by SB334867, an orexin receptor 1 (OXR1) blocker and bisindolylmaleimide, a protein kinase C (PKC) inhibitor, indicating the involvement of OXR1 and PKC. bisindolylmaleimide 89-108 hypocretin Mus musculus 15-23 19915095-6 2010 The excitatory effect of orexin A on PL pyramidal neurons was enhanced when HCN currents were diminished, while attenuated when HCN currents were enlarged. Hydrogen Cyanide 76-79 hypocretin Mus musculus 25-33 19945404-0 2009 Hypothalamic orexin stimulates feeding-associated glucose utilization in skeletal muscle via sympathetic nervous system. Glucose 50-57 hypocretin Mus musculus 13-19 19703479-5 2010 In the present study, locomotor activity in a novel environment and dopamine turnover was significantly decreased in orexin-deficient mice compared to WT mice, which suggests that psychostimulants may be useful for maintaining wakefulness in orexin deficiency. Dopamine 68-76 hypocretin Mus musculus 117-123 19703479-7 2010 The hyperlocomotion induced by methamphetamine and methylphenidate was lower, whereas that induced by MDMA was higher in orexin KO mice compared to WT mice. N-Methyl-3,4-methylenedioxyamphetamine 102-106 hypocretin Mus musculus 121-127 20034477-0 2010 Orexin mediates morphine place preference, but not morphine-induced hyperactivity or sensitization. Morphine 16-24 hypocretin Mus musculus 0-6 20034477-2 2010 Here, we investigated orexin"s contribution to morphine-induced behavioral sensitization and place preference. Morphine 47-55 hypocretin Mus musculus 22-28 20034477-4 2010 C57BL/6J mice received the orexin receptor 1 (Ox1r) antagonist, SB-334867, prior to test sessions. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 64-73 hypocretin Mus musculus 27-33 19489767-3 2010 In addition, a new functional role of orexin is emerging in the regulation of insulin and leptin sensitivities responsible for whole-body glucose metabolism. Glucose 138-145 hypocretin Mus musculus 38-44 19945404-2 2009 We show that injection of orexin-A into the ventromedial hypothalamus (VMH) of mice or rats increased glucose uptake and promoted insulin-induced glucose uptake and glycogen synthesis in skeletal muscle, but not in white adipose tissue, by activating the sympathetic nervous system. Glucose 102-109 hypocretin Mus musculus 26-34 19945404-2 2009 We show that injection of orexin-A into the ventromedial hypothalamus (VMH) of mice or rats increased glucose uptake and promoted insulin-induced glucose uptake and glycogen synthesis in skeletal muscle, but not in white adipose tissue, by activating the sympathetic nervous system. Glucose 146-153 hypocretin Mus musculus 26-34 19945404-2 2009 We show that injection of orexin-A into the ventromedial hypothalamus (VMH) of mice or rats increased glucose uptake and promoted insulin-induced glucose uptake and glycogen synthesis in skeletal muscle, but not in white adipose tissue, by activating the sympathetic nervous system. Glycogen 165-173 hypocretin Mus musculus 26-34 19923277-6 2009 These data indicate that Ox, but not HA, promotes W through enhanced locomotion and suggest that HA and Ox neurons exert a distinct, but complementary and synergistic control of W: the neuropeptide being more involved in its behavioral aspects, whereas the amine is mainly responsible for its qualitative cognitive aspects and cortical EEG activation. Amines 257-262 hypocretin Mus musculus 25-27 19809293-11 2009 Microinjection of the orexin-1 receptor antagonist SB-334867-A during the active period slowed firing rates of locus coeruleus neurons in halothane-anesthetized rats, but had no effect on isoflurane-anesthetized rats. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 51-62 hypocretin Mus musculus 22-28 19923277-6 2009 These data indicate that Ox, but not HA, promotes W through enhanced locomotion and suggest that HA and Ox neurons exert a distinct, but complementary and synergistic control of W: the neuropeptide being more involved in its behavioral aspects, whereas the amine is mainly responsible for its qualitative cognitive aspects and cortical EEG activation. Amines 257-262 hypocretin Mus musculus 104-106 19809293-11 2009 Microinjection of the orexin-1 receptor antagonist SB-334867-A during the active period slowed firing rates of locus coeruleus neurons in halothane-anesthetized rats, but had no effect on isoflurane-anesthetized rats. Halothane 138-147 hypocretin Mus musculus 22-28 19809293-14 2009 Normal emergence from halothane-induced hypnosis in orexin-deficient mice suggests that additional wake-promoting systems likely remain active during general anesthesia produced by halothane. Halothane 22-31 hypocretin Mus musculus 52-58 19750917-1 2009 STUDY OBJECTIVES: The orexin-producing neurons are hypothesized to be essential for the circadian control of sleep/wake behavior, but it remains unknown whether these rhythms are mediated by the orexin peptides or by other signaling molecules released by these neurons such as glutamate or dynorphin. Glutamic Acid 277-286 hypocretin Mus musculus 22-28 19809293-14 2009 Normal emergence from halothane-induced hypnosis in orexin-deficient mice suggests that additional wake-promoting systems likely remain active during general anesthesia produced by halothane. Halothane 181-190 hypocretin Mus musculus 52-58 19623260-4 2009 To identify a potential neuronal circuit, the neurotoxin hypocretin-2-saporin (HCRT2-SAP) was used to lesion neurons in the ventral lateral periaquaductal gray (vlPAG). hcrt2-sap 79-88 hypocretin Mus musculus 57-67 19623260-6 2009 Indeed, HCRT-ko mice (n = 8) given the neurotoxin HCRT2-SAP (16.5 ng/23nl/sec each side) in the vlPAG had levels of REM sleep and sleep fragmentation that were considerably higher compared to HCRT-ko given saline (+39%; n = 7) or wildtype mice (+177%; n = 9). Sodium Chloride 206-212 hypocretin Mus musculus 8-12 19623260-6 2009 Indeed, HCRT-ko mice (n = 8) given the neurotoxin HCRT2-SAP (16.5 ng/23nl/sec each side) in the vlPAG had levels of REM sleep and sleep fragmentation that were considerably higher compared to HCRT-ko given saline (+39%; n = 7) or wildtype mice (+177%; n = 9). Sodium Chloride 206-212 hypocretin Mus musculus 50-54 19442935-0 2009 CO2 activates orexin-containing neurons in mice. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 0-3 hypocretin Mus musculus 14-20 19508695-2 2009 It was recently proposed that TASK (TWIK-related acid-sensitive potassium) channels [TASK1 (K(2P)3.1) and/or TASK3 (K(2P)9.1)] regulate neuronal firing and may contribute to the specialized responses of orexin neurons to glucose and pH. Glucose 221-228 hypocretin Mus musculus 203-209 19508695-6 2009 However, orexin neurons from TASK1/3 knockout mice retained typical responses to glucose and pH, and the knockout animals showed normal food-anticipatory locomotor activity. Glucose 81-88 hypocretin Mus musculus 9-15 19663260-11 2009 It has also been observed in mice with orexin deficit produced by molecular genetics that they have a significantly low development of dependency on amphetamine, a relative compound of methylphenidate. Amphetamine 149-160 hypocretin Mus musculus 39-45 19663260-11 2009 It has also been observed in mice with orexin deficit produced by molecular genetics that they have a significantly low development of dependency on amphetamine, a relative compound of methylphenidate. Methylphenidate 185-200 hypocretin Mus musculus 39-45 19442935-6 2009 Although current methodology cannot differentiate between direct effect of CO(2) on the orexin-containing neurons and indirect one through other neurons, this is the first report showing that inhalation of CO(2) did activate the orexin-containing neurons in vivo. co(2) 206-211 hypocretin Mus musculus 88-94 19442935-6 2009 Although current methodology cannot differentiate between direct effect of CO(2) on the orexin-containing neurons and indirect one through other neurons, this is the first report showing that inhalation of CO(2) did activate the orexin-containing neurons in vivo. co(2) 206-211 hypocretin Mus musculus 229-235 19246384-3 2009 In oxGKO brain slices, we show that the absence of GABA(B) receptors decreases the sensitivity of orexin neurons to both excitatory and inhibitory inputs because of augmented GABA(A)-mediated inhibition that increases the membrane conductance and shunts postsynaptic currents in these neurons. gamma-Aminobutyric Acid 51-55 hypocretin Mus musculus 98-104 19751316-4 2009 The antagonist properties of EMPA were determined by Schild analysis using the orexin-A- or orexin-B-induced accumulation of [(3)H]inositol phosphates (IP). ip 152-154 hypocretin Mus musculus 92-100 19751316-10 2009 EMPA significantly reversed [Ala(11),D-Leu(15)]orexin-B-induced hyperlocomotion dose-dependently during the resting phase in mice. Alanine 29-32 hypocretin Mus musculus 47-55 19751316-10 2009 EMPA significantly reversed [Ala(11),D-Leu(15)]orexin-B-induced hyperlocomotion dose-dependently during the resting phase in mice. D-LEUCINE 37-42 hypocretin Mus musculus 47-55 19273574-1 2009 Recent data from transgenic mice suggest that orexin plays an important role in the ventilatory response to CO(2) during wakefulness. co(2) 108-113 hypocretin Mus musculus 46-52 19751316-4 2009 The antagonist properties of EMPA were determined by Schild analysis using the orexin-A- or orexin-B-induced accumulation of [(3)H]inositol phosphates (IP). [(3)h]inositol phosphates 125-150 hypocretin Mus musculus 92-100 19321767-8 2009 In contrast, TRH significantly reduced the frequency, but not amplitude, of miniature excitatory PSCs without affecting miniature inhibitory PSC frequency or amplitude, indicating that TRH also reduces the probability of glutamate release onto Hcrt neurons. Glutamic Acid 221-230 hypocretin Mus musculus 244-248 19246384-4 2009 This increase in GABA(A)-mediated inhibitory tone is apparently the result of an orexin receptor type 1-mediated activation of local GABAergic interneurons that project back onto orexin neurons. gamma-Aminobutyric Acid 17-21 hypocretin Mus musculus 81-87 19084548-11 2009 UCP1 (p<0.01) and UCP3 (p<0.05) mRNA expressions were greater in risperidone-treated mice compared to controls, whereas, orexin mRNA expression was lower in risperidone-treated mice (p<0.01). Risperidone 163-174 hypocretin Mus musculus 127-133 19084548-12 2009 These results suggest that risperidone-induced weight gain in mice is a consequence of increased energy intake and reduced activity, while the elevation in body temperature may be a result of thermogenic effect of food intake and elevated UCP1, UCP3, and a reduced hypothalamic orexin expression. Risperidone 27-38 hypocretin Mus musculus 278-284 18716204-2 2008 Although the compound is reported to affect multiple neurotransmitter systems such as catecholamines, serotonin, glutamate, GABA, orexin, and histamine, however, the molecular mechanism by which modafinil increases wakefulness is debated. Modafinil 195-204 hypocretin Mus musculus 130-136 19193897-8 2009 Hypocretin also enhanced excitatory inputs to POMC cells via a presynaptic mechanism and indirectly increased the release of GABA onto these cells in a spike-dependent manner. gamma-Aminobutyric Acid 125-129 hypocretin Mus musculus 0-10 18952152-12 2008 Moreover, the OX-A-induced increase in neuropeptide Y (NPY)-positive cells in the hilus of the dentate gyrus was blocked by SB-334867. Antimony 124-126 hypocretin Mus musculus 14-18 18999901-5 2008 Fos/Hcrt co-localizations were processed by employing avidin-biotin-peroxidase (ABC) complex and diaminobenzidine chromogen for Hcrt labeling. 4,4'-Dihydrazino-biphenyl 97-113 hypocretin Mus musculus 4-8 18828950-6 2008 Knockout of the orexin gene in mice reduces CO2-induced increases in breathing by approximately 50% and increases the frequency of spontaneous sleep apneas. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 44-47 hypocretin Mus musculus 16-22 18828950-7 2008 The relationship between orexins and breathing may be bidirectional: the rate of breathing controls acid and CO2 levels, and these signals alter the electrical activity of orexin neurons in vitro. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 109-112 hypocretin Mus musculus 25-31 18591392-0 2008 Metabolism-independent sugar sensing in central orexin neurons. Sugars 23-28 hypocretin Mus musculus 48-54 18591392-3 2008 Orexin/hypocretin neurons of the lateral hypothalamus are widely projecting glucose-inhibited cells essential for normal cognitive arousal and feeding behavior. Glucose 76-83 hypocretin Mus musculus 0-6 18591392-3 2008 Orexin/hypocretin neurons of the lateral hypothalamus are widely projecting glucose-inhibited cells essential for normal cognitive arousal and feeding behavior. Glucose 76-83 hypocretin Mus musculus 7-17 18591392-4 2008 Here, we used different sugars, energy metabolites, and pharmacological tools to explore the glucose-sensing strategy of orexin cells. Sugars 24-30 hypocretin Mus musculus 121-127 18591392-4 2008 Here, we used different sugars, energy metabolites, and pharmacological tools to explore the glucose-sensing strategy of orexin cells. Glucose 93-100 hypocretin Mus musculus 121-127 18295497-3 2008 In narcoleptic orexin(-/-) mice, tiprolisant enhanced HA and noradrenaline neuronal activity, promoted wakefulness and decreased abnormal DREMs, all effects being amplified by co-administration of modafinil, a currently-prescribed wake-promoting drug. pitolisant 33-44 hypocretin Mus musculus 15-21 18198212-2 2008 We report here that orexin-induced apoptosis is driven by an immunoreceptor tyrosine-based inhibitory motif (ITIM) (IIY(358)NFL) present in the OX1R. Tyrosine 76-84 hypocretin Mus musculus 20-26 18395341-5 2008 Likewise, in the mouse adrenal medulla slices, orexin A also induced catecholamine release mainly through the activation of OX1R. Catecholamines 69-82 hypocretin Mus musculus 47-55 18423425-0 2008 Orexin mediates the expression of precipitated morphine withdrawal and concurrent activation of the nucleus accumbens shell. Morphine 47-55 hypocretin Mus musculus 0-6 18423425-3 2008 We investigated the mechanism and neuroanatomic basis of orexin"s role in morphine withdrawal. Morphine 74-82 hypocretin Mus musculus 57-63 18423425-12 2008 CONCLUSIONS: Altogether, these data demonstrate that orexin, acting via Ox1r, is critical for the expression of morphine withdrawal. Morphine 112-120 hypocretin Mus musculus 53-59 18423425-13 2008 AcbSh activation during withdrawal is dependent on Ox1r function and is likely mediated by indirect action of LH orexin neurons. acbsh 0-5 hypocretin Mus musculus 113-119 18551194-6 2008 N/OFQ also modulated presynaptic release of GABA and glutamate onto Hcrt neurons in mouse hypothalamic slices. gamma-Aminobutyric Acid 44-48 hypocretin Mus musculus 68-72 18551194-6 2008 N/OFQ also modulated presynaptic release of GABA and glutamate onto Hcrt neurons in mouse hypothalamic slices. Glutamic Acid 53-62 hypocretin Mus musculus 68-72 18256806-3 2008 METHODS: Orexin knockout mice fasted overnight underwent oral glucose tolerance testing and insulin tolerance testing. Glucose 62-69 hypocretin Mus musculus 9-15 16959056-4 2007 Moreover, CP 94253 (5-10 mg/kg), a selective 5-HT1B receptor agonist, exerted appetite-suppressing effects and significantly increased hypothalamic pro-opiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcript (CART) gene expression and decreased hypothalamic orexin gene expression. CP 94253 10-18 hypocretin Mus musculus 276-282 18344611-2 2008 In the present study, we showed that a muscarinic receptor agonist, carbachol (CCh), activates almost 20% of orexin-producing neurons (orexin neurons), which play a critical role in maintenance of arousal. Carbachol 68-77 hypocretin Mus musculus 109-115 18344611-2 2008 In the present study, we showed that a muscarinic receptor agonist, carbachol (CCh), activates almost 20% of orexin-producing neurons (orexin neurons), which play a critical role in maintenance of arousal. Carbachol 68-77 hypocretin Mus musculus 135-141 18344611-2 2008 In the present study, we showed that a muscarinic receptor agonist, carbachol (CCh), activates almost 20% of orexin-producing neurons (orexin neurons), which play a critical role in maintenance of arousal. Carbachol 79-82 hypocretin Mus musculus 109-115 18344611-2 2008 In the present study, we showed that a muscarinic receptor agonist, carbachol (CCh), activates almost 20% of orexin-producing neurons (orexin neurons), which play a critical role in maintenance of arousal. Carbachol 79-82 hypocretin Mus musculus 135-141 18344611-3 2008 We also found that a very small population of orexin neurons (1%) was inhibited by CCh. Carbachol 83-86 hypocretin Mus musculus 46-52 18344611-4 2008 Muscarinic receptor antagonists inhibited the CCh-induced activation of orexin neurons in a dose-dependent manner. Carbachol 46-49 hypocretin Mus musculus 72-78 18344611-8 2008 These results indicate that CCh activates 20% of orexin neurons through the M(3) muscarinic receptor and subsequent activation of nonselective cation channels. Carbachol 28-31 hypocretin Mus musculus 49-55 18032578-2 2008 Raphe nuclei, which modulate several physiological functions through serotonin, receive dense projections from orexin-containing neurons in the hypothalamus. Serotonin 69-78 hypocretin Mus musculus 111-117 18025760-9 2007 When JVS mice were fed a carnitine-rich diet, suppression of expression of orexin mRNA in hypothalamus was eliminated, and on day 28 lactose and cellulose were detected in the stomach without hypoglycemia. Carnitine 25-34 hypocretin Mus musculus 75-81 18060037-4 2007 Here, we show that acute and chronic prolonged wakefulness in mice induced by modafinil treatment produced long-term potentiation (LTP) of glutamatergic synapses on hypocretin/orexin neurons in the lateral hypothalamus, a well-established arousal/wake-promoting center. Modafinil 78-87 hypocretin Mus musculus 165-175 18171940-7 2008 Subsequently activation of phospholipase Cbeta triggers an increase in intracellular calcium by both calcium influx through nonselective cation channels and calcium release from calcium stores in orexin neurons. Calcium 85-92 hypocretin Mus musculus 196-202 16959056-5 2007 These results suggest that fluvoxamine and inactivation of 5-HT2C receptors exert feeding suppression through activation of 5-HT1B receptors, and that 5-HT1B receptors up-regulate hypothalamic POMC and CART gene expression and down-regulate hypothalamic orexin gene expression in mice. Fluvoxamine 27-38 hypocretin Mus musculus 254-260 17680885-5 2007 Orexin expression in the lateral hypothalamus was significantly increased in corticosterone deficient Pomc(-/-) mice. Corticosterone 77-91 hypocretin Mus musculus 0-6 17717124-9 2007 Supplementation of orexin-A or -B (3 nmol) partially restored the hypercapnic chemoreflex in ORX-KO mice (0.28 +/- 0.03 mlxmin(-1).g(-1)x% CO(2)(-1) for orexin-A and 0.32 +/- 0.04 mlxmin(-1)xg(-1)x% CO(2)(-1) for orexin-B). co(2) 139-144 hypocretin Mus musculus 19-33 17717124-9 2007 Supplementation of orexin-A or -B (3 nmol) partially restored the hypercapnic chemoreflex in ORX-KO mice (0.28 +/- 0.03 mlxmin(-1).g(-1)x% CO(2)(-1) for orexin-A and 0.32 +/- 0.04 mlxmin(-1)xg(-1)x% CO(2)(-1) for orexin-B). co(2) 199-204 hypocretin Mus musculus 19-33 17717124-12 2007 Our findings suggest that orexin plays a crucial role in CO(2) sensitivity at least during wake periods in mice. co(2) 57-62 hypocretin Mus musculus 26-32 17475795-1 2007 Cannabinoids modulate energy homeostasis and decrease cognitive arousal, possibly by acting on hypothalamic neurons including those that synthesize melanin-concentrating hormone (MCH) or hypocretin/orexin. Cannabinoids 0-12 hypocretin Mus musculus 187-204 16849632-9 2006 We hypothesize that abnormal hypothalamic orexin expression leads to changes in liver carbohydrate metabolism and may contribute to the moderate obesity observed in tubby mice. Carbohydrates 86-98 hypocretin Mus musculus 42-48 16959906-9 2007 Our findings suggest that orexin plays a crucial role both in CO(2) sensitivity during wakefulness and in preserving ventilation stability during sleep. co(2) 62-67 hypocretin Mus musculus 26-32 17093123-0 2007 Adenosine inhibits activity of hypocretin/orexin neurons by the A1 receptor in the lateral hypothalamus: a possible sleep-promoting effect. Adenosine 0-9 hypocretin Mus musculus 31-48 17093123-6 2007 In this study, we examined the hypothesis that adenosine inhibits the activity of hypocretin/orexin neurons by using electrophysiological methods in brain slices from mice expressing green fluorescent protein in hypocretin/orexin neurons. Adenosine 47-56 hypocretin Mus musculus 82-99 17093123-6 2007 In this study, we examined the hypothesis that adenosine inhibits the activity of hypocretin/orexin neurons by using electrophysiological methods in brain slices from mice expressing green fluorescent protein in hypocretin/orexin neurons. Adenosine 47-56 hypocretin Mus musculus 212-229 17093123-8 2007 The adenosine-mediated inhibition arises from depression of excitatory synaptic transmission to hypocretin/orexin neurons because adenosine depresses the amplitude of evoked excitatory postsynaptic potential and the frequency of spontaneous and miniature excitatory postsynaptic currents in these neurons. Adenosine 4-13 hypocretin Mus musculus 96-113 17093123-8 2007 The adenosine-mediated inhibition arises from depression of excitatory synaptic transmission to hypocretin/orexin neurons because adenosine depresses the amplitude of evoked excitatory postsynaptic potential and the frequency of spontaneous and miniature excitatory postsynaptic currents in these neurons. Adenosine 130-139 hypocretin Mus musculus 96-113 17093123-9 2007 At the cell body of the hypocretin/orexin neurons, adenosine inhibits voltage-dependent calcium currents without the induction of GIRK current. Adenosine 51-60 hypocretin Mus musculus 24-41 17093123-11 2007 In summary, our data suggest that in addition to its effect in the basal forebrain, adenosine exerts its sleep-promoting effect in the LH by inhibition of hypocretin/orexin neurons. Adenosine 84-93 hypocretin Mus musculus 155-172 16872646-5 2006 Orexin-A increased the histamine release and locomotor activity, but not food intake, suggesting that the histaminergic system participates in arousal rather than feeding by orexin-A. Histamine 23-32 hypocretin Mus musculus 0-8 16867176-7 2006 Intraperitoneal injections of the HCRT-1 receptor antagonist, SB-334867, exhibited a general trend towards increasing time spent low-arched back nursing (P = 0.053) and decreasing licking and grooming of pups while high-arched back nursing (P = 0.052). 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 62-71 hypocretin Mus musculus 34-38 16556901-3 2006 Because orexin is released during wakefulness and is thought to promote heat production, we hypothesized that orexin KO mice would have lower Tb while awake. Terbium 142-144 hypocretin Mus musculus 8-14 16556901-3 2006 Because orexin is released during wakefulness and is thought to promote heat production, we hypothesized that orexin KO mice would have lower Tb while awake. Terbium 142-144 hypocretin Mus musculus 110-116 16556901-5 2006 Orexin KO mice had normal diurnal variations in Tb, but the ultradian rhythms of Tb, locomotor activity, and wakefulness were markedly reduced. Terbium 48-50 hypocretin Mus musculus 0-6 16762378-6 2006 Exogenous administration of either OXA or nicotine induced a transient contraction that was completely inhibited by atropine and tetrodotoxin. Atropine 116-124 hypocretin Mus musculus 35-38 16762378-6 2006 Exogenous administration of either OXA or nicotine induced a transient contraction that was completely inhibited by atropine and tetrodotoxin. Tetrodotoxin 129-141 hypocretin Mus musculus 35-38 16762378-7 2006 The OXA-induced contraction was significantly inhibited by hexamethonium and SB-334867-A, whereas the nicotine-induced contraction was not inhibited by SB-334867-A. Hexamethonium 59-72 hypocretin Mus musculus 4-7 16762378-7 2006 The OXA-induced contraction was significantly inhibited by hexamethonium and SB-334867-A, whereas the nicotine-induced contraction was not inhibited by SB-334867-A. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 77-86 hypocretin Mus musculus 4-7 16762378-11 2006 Exogenous OXA, in the presence of atropine, induced a relaxation that was significantly inhibited by both l-nitroarginine and SB-334867-A, but not by hexamethonium. Atropine 34-42 hypocretin Mus musculus 10-13 16762378-11 2006 Exogenous OXA, in the presence of atropine, induced a relaxation that was significantly inhibited by both l-nitroarginine and SB-334867-A, but not by hexamethonium. H-Arg(NO2)-OH 106-121 hypocretin Mus musculus 10-13 16762378-11 2006 Exogenous OXA, in the presence of atropine, induced a relaxation that was significantly inhibited by both l-nitroarginine and SB-334867-A, but not by hexamethonium. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 126-137 hypocretin Mus musculus 10-13 16611835-0 2006 Orexin neurons are directly and indirectly regulated by catecholamines in a complex manner. Catecholamines 56-70 hypocretin Mus musculus 0-6 16611835-2 2006 In the present study, we show that NA, dopamine, and adrenaline all directly hyperpolarized orexin neurons. Dopamine 39-47 hypocretin Mus musculus 92-98 16627567-8 2006 The existence of GABA(B) modulation was supported by GABA(B(1)) subunit immunoreactivity on Hcrt cells colabelled with antisera to the Hcrt-2 peptide. gamma-Aminobutyric Acid 17-21 hypocretin Mus musculus 92-96 16627567-8 2006 The existence of GABA(B) modulation was supported by GABA(B(1)) subunit immunoreactivity on Hcrt cells colabelled with antisera to the Hcrt-2 peptide. gamma-Aminobutyric Acid 17-21 hypocretin Mus musculus 135-139 16627567-11 2006 These data suggest that GABA(B) receptors modulate Hcrt neuronal activity via both pre- and postsynaptic mechanisms, which may underlie the promotion of non-rapid eye movement sleep and have implications for the use of GABA(B) agonists in the treatment of substance addiction through direct interaction with the Hcrt system. gamma-Aminobutyric Acid 24-28 hypocretin Mus musculus 51-55 16627567-11 2006 These data suggest that GABA(B) receptors modulate Hcrt neuronal activity via both pre- and postsynaptic mechanisms, which may underlie the promotion of non-rapid eye movement sleep and have implications for the use of GABA(B) agonists in the treatment of substance addiction through direct interaction with the Hcrt system. gamma-Aminobutyric Acid 24-28 hypocretin Mus musculus 312-316 16611835-2 2006 In the present study, we show that NA, dopamine, and adrenaline all directly hyperpolarized orexin neurons. Epinephrine 53-63 hypocretin Mus musculus 92-98 16611835-1 2006 We reported elsewhere that orexin neurons are directly hyperpolarized by noradrenaline (NA) and dopamine. Norepinephrine 73-86 hypocretin Mus musculus 27-33 16611835-6 2006 This response was inhibited by alpha1-AR antagonist, prazosin, which suggests the existence of alpha1-ARs on the orexin neurons along with alpha2-AR. Prazosin 53-61 hypocretin Mus musculus 113-119 16611835-11 2006 The evidence presented here revealed that orexin neurons are regulated by catecholamines in a complex manner. Catecholamines 74-88 hypocretin Mus musculus 42-48 16611835-1 2006 We reported elsewhere that orexin neurons are directly hyperpolarized by noradrenaline (NA) and dopamine. Dopamine 96-104 hypocretin Mus musculus 27-33 16410401-11 2006 The present study provided further support for our hypothesis that orexin-containing neurons in PFA play a role as a master switch to activate multiple efferent pathways of the defense response and also operate as a regulator of basal AP. pfa 96-99 hypocretin Mus musculus 67-73 16492946-10 2006 The robust excitation evoked by Hcrt-2 on cortically projecting glutamate PVT neurons could generate substantial excitation in multiple layers of the mPFC, adding to the more selective direct excitatory actions of Hcrt in the mPFC and potentially increasing cortical arousal and attention to limbic or visceral states. Glutamic Acid 64-73 hypocretin Mus musculus 32-36 16484324-11 2006 Des-acyl ghrelin increased the intracellular calcium concentrations in isolated orexin neurons. Calcium 45-52 hypocretin Mus musculus 80-86 16492946-10 2006 The robust excitation evoked by Hcrt-2 on cortically projecting glutamate PVT neurons could generate substantial excitation in multiple layers of the mPFC, adding to the more selective direct excitatory actions of Hcrt in the mPFC and potentially increasing cortical arousal and attention to limbic or visceral states. Glutamic Acid 64-73 hypocretin Mus musculus 214-218 15958604-0 2005 Direct excitation of hypocretin/orexin cells by extracellular ATP at P2X receptors. Adenosine Triphosphate 62-65 hypocretin Mus musculus 21-31 17192702-6 2006 Administration of 1 microM OX1R antagonist, SB-334867, completely blocked the observed orexin A responses in these cells, indicating that orexin A stimulation of GnRH neurons is specifically through OX1R. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 44-53 hypocretin Mus musculus 87-95 17192702-6 2006 Administration of 1 microM OX1R antagonist, SB-334867, completely blocked the observed orexin A responses in these cells, indicating that orexin A stimulation of GnRH neurons is specifically through OX1R. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 44-53 hypocretin Mus musculus 138-146 16321792-8 2005 In the mouse lateral hypothalamus, orexin/hypocretin neurons (which promote wakefulness, locomotor activity and foraging) are glucose-inhibited, whereas melanin-concentrating hormone neurons (which promote sleep and energy conservation) are glucose-excited. Glucose 126-133 hypocretin Mus musculus 35-41 16321792-8 2005 In the mouse lateral hypothalamus, orexin/hypocretin neurons (which promote wakefulness, locomotor activity and foraging) are glucose-inhibited, whereas melanin-concentrating hormone neurons (which promote sleep and energy conservation) are glucose-excited. Glucose 241-248 hypocretin Mus musculus 35-41 16202530-2 2005 Orexin neurons projected to the tuberomammillary nucleus and orexins may release histamine from the histamine neurons in this nucleus. Histamine 81-90 hypocretin Mus musculus 0-6 16202530-2 2005 Orexin neurons projected to the tuberomammillary nucleus and orexins may release histamine from the histamine neurons in this nucleus. Histamine 100-109 hypocretin Mus musculus 0-6 16202530-5 2005 Here we studied the effects of histamine H1 and H2 receptors on orexin A-produced antinociception using histamine receptor knockout mice in four assays of nociception; the hot-plate, the tail-flick, the tail-pressure and the capsaicin tests. Capsaicin 225-234 hypocretin Mus musculus 64-72 16202530-12 2005 These findings suggest the possibility that orexin A may activate H1 and H2 receptors in the supraspinal levels through the release of histamine from neurons, which might attenuate the antinociceptive effects of orexin A. Histamine 135-144 hypocretin Mus musculus 44-52 16202530-12 2005 These findings suggest the possibility that orexin A may activate H1 and H2 receptors in the supraspinal levels through the release of histamine from neurons, which might attenuate the antinociceptive effects of orexin A. Histamine 135-144 hypocretin Mus musculus 212-220 16407535-3 2006 The levels of dopamine and its major metabolites in the nucleus accumbens were markedly increased by the microinjection of orexin A and orexin B into the VTA. Dopamine 14-22 hypocretin Mus musculus 123-131 16407535-3 2006 The levels of dopamine and its major metabolites in the nucleus accumbens were markedly increased by the microinjection of orexin A and orexin B into the VTA. Dopamine 14-22 hypocretin Mus musculus 136-144 16407535-4 2006 The subcutaneous morphine-induced place preference and hyperlocomotion observed in wild-type mice were abolished in mice that lacked the prepro-orexin gene. Morphine 17-25 hypocretin Mus musculus 137-150 16407535-5 2006 An intra-VTA injection of a selective orexin receptor antagonist SB334867A [1-(2-methylbenzoxazol-6-yl)-3-[1.5]naphthyridin-4-yl urea] significantly suppressed the morphine-induced place preference in rats. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 65-74 hypocretin Mus musculus 38-44 16407535-5 2006 An intra-VTA injection of a selective orexin receptor antagonist SB334867A [1-(2-methylbenzoxazol-6-yl)-3-[1.5]naphthyridin-4-yl urea] significantly suppressed the morphine-induced place preference in rats. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 76-133 hypocretin Mus musculus 38-44 16407535-6 2006 Furthermore, the increased level of dialysate dopamine produced by morphine in the mouse brain was significantly decreased by deletion of the prepro-orexin gene. Dopamine 46-54 hypocretin Mus musculus 142-155 16407535-6 2006 Furthermore, the increased level of dialysate dopamine produced by morphine in the mouse brain was significantly decreased by deletion of the prepro-orexin gene. Morphine 67-75 hypocretin Mus musculus 142-155 16407535-7 2006 These findings provide new evidence that orexin-containing neurons in the VTA are directly implicated in the rewarding effect and hyperlocomotion induced by morphine through activation of the mesolimbic dopamine pathway in rodents. Morphine 157-165 hypocretin Mus musculus 41-47 16407535-7 2006 These findings provide new evidence that orexin-containing neurons in the VTA are directly implicated in the rewarding effect and hyperlocomotion induced by morphine through activation of the mesolimbic dopamine pathway in rodents. Dopamine 203-211 hypocretin Mus musculus 41-47 15896493-7 2005 Pharmacological inhibition of the synthesis of epinephrine, a potential neurotransmitter at central motoric alpha1-adrenoceptors, with 2,3-dichloro-alpha-methylbenzylamine also significantly attenuated orexin A-induced hyperactivity. Epinephrine 47-58 hypocretin Mus musculus 202-210 16093397-6 2005 The CCKA receptor antagonist lorglumide inhibited CCK-8S-induced activation of orexin neurons, whereas the CCKB receptor agonists CCK-4 (a tetrapeptide form of cholecystokinin) and nonsulfated CCK-8 had little effect. lorglumide 29-39 hypocretin Mus musculus 79-85 15896493-7 2005 Pharmacological inhibition of the synthesis of epinephrine, a potential neurotransmitter at central motoric alpha1-adrenoceptors, with 2,3-dichloro-alpha-methylbenzylamine also significantly attenuated orexin A-induced hyperactivity. 2,3-dichloro-alpha-methylbenzylamine 135-171 hypocretin Mus musculus 202-210 15808907-12 2005 The effects of orexin A were completely blocked by adenosine type 1 receptor antagonists, 1,3-dipropyl-8-cyclopentylxanthine (DPCPX) and theophylline, but not by naloxone, suggesting a possible involvement of the adenosine-containing neurons and/or the adenosine pathway in these orexin actions. Adenosine 51-60 hypocretin Mus musculus 15-21 15808907-12 2005 The effects of orexin A were completely blocked by adenosine type 1 receptor antagonists, 1,3-dipropyl-8-cyclopentylxanthine (DPCPX) and theophylline, but not by naloxone, suggesting a possible involvement of the adenosine-containing neurons and/or the adenosine pathway in these orexin actions. Adenosine 213-222 hypocretin Mus musculus 15-23 15808906-0 2005 Orexin-A-induced feeding is dependent on nitric oxide. Nitric Oxide 41-53 hypocretin Mus musculus 0-8 15808906-6 2005 We next examined the effects of Nomega-nitro-L-arginine methyl ester (L-NAME), a nitric oxide synthase inhibitor, on orexin-A-induced increase in food intake. NG-Nitroarginine Methyl Ester 32-68 hypocretin Mus musculus 117-125 15808906-6 2005 We next examined the effects of Nomega-nitro-L-arginine methyl ester (L-NAME), a nitric oxide synthase inhibitor, on orexin-A-induced increase in food intake. NG-Nitroarginine Methyl Ester 70-76 hypocretin Mus musculus 117-125 15808906-7 2005 L-NAME (50 mg/kg; SC) significantly blocked the orexin-A-induced increase in food intake. NG-Nitroarginine Methyl Ester 0-6 hypocretin Mus musculus 48-56 15808907-12 2005 The effects of orexin A were completely blocked by adenosine type 1 receptor antagonists, 1,3-dipropyl-8-cyclopentylxanthine (DPCPX) and theophylline, but not by naloxone, suggesting a possible involvement of the adenosine-containing neurons and/or the adenosine pathway in these orexin actions. 1,3-dipropyl-8-cyclopentylxanthine 90-124 hypocretin Mus musculus 15-23 15808907-12 2005 The effects of orexin A were completely blocked by adenosine type 1 receptor antagonists, 1,3-dipropyl-8-cyclopentylxanthine (DPCPX) and theophylline, but not by naloxone, suggesting a possible involvement of the adenosine-containing neurons and/or the adenosine pathway in these orexin actions. 1,3-dipropyl-8-cyclopentylxanthine 90-124 hypocretin Mus musculus 15-21 15808907-12 2005 The effects of orexin A were completely blocked by adenosine type 1 receptor antagonists, 1,3-dipropyl-8-cyclopentylxanthine (DPCPX) and theophylline, but not by naloxone, suggesting a possible involvement of the adenosine-containing neurons and/or the adenosine pathway in these orexin actions. 1,3-dipropyl-8-cyclopentylxanthine 126-131 hypocretin Mus musculus 15-23 15808907-12 2005 The effects of orexin A were completely blocked by adenosine type 1 receptor antagonists, 1,3-dipropyl-8-cyclopentylxanthine (DPCPX) and theophylline, but not by naloxone, suggesting a possible involvement of the adenosine-containing neurons and/or the adenosine pathway in these orexin actions. 1,3-dipropyl-8-cyclopentylxanthine 126-131 hypocretin Mus musculus 15-21 15808907-12 2005 The effects of orexin A were completely blocked by adenosine type 1 receptor antagonists, 1,3-dipropyl-8-cyclopentylxanthine (DPCPX) and theophylline, but not by naloxone, suggesting a possible involvement of the adenosine-containing neurons and/or the adenosine pathway in these orexin actions. Theophylline 137-149 hypocretin Mus musculus 15-23 15808907-12 2005 The effects of orexin A were completely blocked by adenosine type 1 receptor antagonists, 1,3-dipropyl-8-cyclopentylxanthine (DPCPX) and theophylline, but not by naloxone, suggesting a possible involvement of the adenosine-containing neurons and/or the adenosine pathway in these orexin actions. Adenosine 213-222 hypocretin Mus musculus 15-21 15808907-12 2005 The effects of orexin A were completely blocked by adenosine type 1 receptor antagonists, 1,3-dipropyl-8-cyclopentylxanthine (DPCPX) and theophylline, but not by naloxone, suggesting a possible involvement of the adenosine-containing neurons and/or the adenosine pathway in these orexin actions. Theophylline 137-149 hypocretin Mus musculus 15-21 15848807-5 2005 Monoamine-containing groups that are innervated by orexin neurons do not receive reciprocal connections, while cholinergic neurons in the basal forebrain have reciprocal connections, which might be important for consolidating wakefulness. monoamine 0-9 hypocretin Mus musculus 51-57 15808907-12 2005 The effects of orexin A were completely blocked by adenosine type 1 receptor antagonists, 1,3-dipropyl-8-cyclopentylxanthine (DPCPX) and theophylline, but not by naloxone, suggesting a possible involvement of the adenosine-containing neurons and/or the adenosine pathway in these orexin actions. Adenosine 51-60 hypocretin Mus musculus 15-23 15848807-6 2005 Electrophysiological study showed that carbachol excites almost one-third of orexin neurons and inhibits a small population of orexin neurons. Carbachol 39-48 hypocretin Mus musculus 77-83 15848807-6 2005 Electrophysiological study showed that carbachol excites almost one-third of orexin neurons and inhibits a small population of orexin neurons. Carbachol 39-48 hypocretin Mus musculus 127-133 15652995-0 2005 Modafinil more effectively induces wakefulness in orexin-null mice than in wild-type littermates. Modafinil 0-9 hypocretin Mus musculus 50-56 15745970-0 2005 Physiological changes in glucose differentially modulate the excitability of hypothalamic melanin-concentrating hormone and orexin neurons in situ. Glucose 25-32 hypocretin Mus musculus 124-130 15745970-5 2005 We also demonstrate that the same physiological shifts in glucose have the opposite effects on the electrical activity of orexin neurons. Glucose 58-65 hypocretin Mus musculus 122-128 15663891-0 2005 Orexin A promotes histamine, but not norepinephrine or serotonin, release in frontal cortex of mice. Histamine 18-27 hypocretin Mus musculus 0-8 15663891-1 2005 AIM: To investigate the effects of orexin A on release of histamine, norepinephrine, and serotonin in the frontal cortex of mice. Histamine 58-67 hypocretin Mus musculus 35-43 15663891-1 2005 AIM: To investigate the effects of orexin A on release of histamine, norepinephrine, and serotonin in the frontal cortex of mice. Norepinephrine 69-83 hypocretin Mus musculus 35-43 15663891-1 2005 AIM: To investigate the effects of orexin A on release of histamine, norepinephrine, and serotonin in the frontal cortex of mice. Serotonin 89-98 hypocretin Mus musculus 35-43 15663891-4 2005 RESULTS: Intracrebroventricular injection of orexin A at doses of 12.5, 50, and 200 pmol per mouse promoted histamine release from the frontal cortex in a dose-dependent manner. Histamine 108-117 hypocretin Mus musculus 45-53 15663891-5 2005 At the highest dose given, 200 pmol, orexin A significantly induced histamine release, with the maximal magnitude being 230% over the mean basal release. Histamine 68-77 hypocretin Mus musculus 37-45 15663891-8 2005 CONCLUSION: These results suggest that the arousal effect of orexin A is mainly mediated by histamine, not by norepinephrine or serotonin. Histamine 92-101 hypocretin Mus musculus 61-69 15663891-8 2005 CONCLUSION: These results suggest that the arousal effect of orexin A is mainly mediated by histamine, not by norepinephrine or serotonin. Norepinephrine 110-124 hypocretin Mus musculus 61-69 15634779-0 2005 Direct and indirect inhibition by catecholamines of hypocretin/orexin neurons. Catecholamines 34-48 hypocretin Mus musculus 52-62 15634779-12 2005 These data suggest that catecholamines evoke strong inhibitory actions on hypocretin neurons and suggest negative feedback from catecholamine cells that may be excited by hypocretin. Catecholamines 24-38 hypocretin Mus musculus 74-84 15634779-12 2005 These data suggest that catecholamines evoke strong inhibitory actions on hypocretin neurons and suggest negative feedback from catecholamine cells that may be excited by hypocretin. Catecholamines 24-37 hypocretin Mus musculus 74-84 15652995-3 2005 Fos protein immunohistochemistry has previously demonstrated that orexin neurons are activated after modafinil administration, and it has been hypothesized that the wakefulness-promoting properties of modafinil might therefore be mediated by the neuropeptide. Modafinil 101-110 hypocretin Mus musculus 66-72 15652995-3 2005 Fos protein immunohistochemistry has previously demonstrated that orexin neurons are activated after modafinil administration, and it has been hypothesized that the wakefulness-promoting properties of modafinil might therefore be mediated by the neuropeptide. Modafinil 201-210 hypocretin Mus musculus 66-72 15652995-7 2005 Surprisingly, modafinil more effectively increased wakefulness time in orexin-/- mice than in the wild-type mice. Modafinil 14-23 hypocretin Mus musculus 71-77 15652995-11 2005 Modafinil administration only partly compensated for this attention deficit in the orexin null mice. Modafinil 0-9 hypocretin Mus musculus 83-89 14999052-0 2004 Hypocretin/orexin peptide signaling in the ascending arousal system: elevation of intracellular calcium in the mouse dorsal raphe and laterodorsal tegmentum. Calcium 96-103 hypocretin Mus musculus 11-17 15256537-9 2004 Although, orexin-A and -B showed no difference in binding characteristics between the splice variants; interestingly, orexin-B led to an increase in IP(3) production at all concentrations in the mOX2 beta R variant. Inositol 1,4,5-Trisphosphate 149-154 hypocretin Mus musculus 118-126 15306649-5 2004 A 5-HT1A receptor agonist, 8-hydroxy-2-(dl-N-propyl-amino)tetralin, also evoked hyperpolarization on orexin neurons with potency comparable with 5-HT. 8-hydroxy-2-(dl-n-propyl-amino)tetralin 27-66 hypocretin Mus musculus 101-107 15306649-9 2004 Intracerebroventricular injection of the 5-HT1A receptor-selective antagonist WAY100635 (100 ng) increased locomotor activity during the latter half of dark phase in wild-type mice but not in orexin/ataxin-3 mice in which orexin neurons are specifically ablated, suggesting that activation of orexin neurons is necessary for the WAY100635-induced increase in locomotor activity. N-(2-(4-(2-methoxyphenyl)-1-piperazinyl)ethyl)-N-(2-pyridinyl)cyclohexanecarboxamide 78-87 hypocretin Mus musculus 222-228 15306649-9 2004 Intracerebroventricular injection of the 5-HT1A receptor-selective antagonist WAY100635 (100 ng) increased locomotor activity during the latter half of dark phase in wild-type mice but not in orexin/ataxin-3 mice in which orexin neurons are specifically ablated, suggesting that activation of orexin neurons is necessary for the WAY100635-induced increase in locomotor activity. N-(2-(4-(2-methoxyphenyl)-1-piperazinyl)ethyl)-N-(2-pyridinyl)cyclohexanecarboxamide 78-87 hypocretin Mus musculus 222-228 14999052-3 2004 Since Hcrt/Orx mobilizes intracellular calcium ([Ca(2+)](i)) in cells transfected with orexin receptors and since receptor-mediated Ca(2+) transients are ubiquitous signaling mechanisms, we investigated whether Hcrt/Orx regulates [Ca(2+)](i) in the LDT and DR. Changes in [Ca(2+)](i) were monitored by fluorescence changes of fura-2 AM loaded cells in young mouse brain slices. Calcium 39-46 hypocretin Mus musculus 6-10 14999052-3 2004 Since Hcrt/Orx mobilizes intracellular calcium ([Ca(2+)](i)) in cells transfected with orexin receptors and since receptor-mediated Ca(2+) transients are ubiquitous signaling mechanisms, we investigated whether Hcrt/Orx regulates [Ca(2+)](i) in the LDT and DR. Changes in [Ca(2+)](i) were monitored by fluorescence changes of fura-2 AM loaded cells in young mouse brain slices. Calcium 39-46 hypocretin Mus musculus 87-93 14999052-7 2004 In contrast, Hcrt/Orx responses were strongly attenuated by lowering extracellular Ca(2+) ( approximately 20 microM) but were not inhibited by concentrations of KB-R7943 (10 microM) selective for blockade of sodium/calcium exchange. orx 18-21 hypocretin Mus musculus 13-17 14999052-7 2004 In contrast, Hcrt/Orx responses were strongly attenuated by lowering extracellular Ca(2+) ( approximately 20 microM) but were not inhibited by concentrations of KB-R7943 (10 microM) selective for blockade of sodium/calcium exchange. Sodium 208-214 hypocretin Mus musculus 13-17 14999052-7 2004 In contrast, Hcrt/Orx responses were strongly attenuated by lowering extracellular Ca(2+) ( approximately 20 microM) but were not inhibited by concentrations of KB-R7943 (10 microM) selective for blockade of sodium/calcium exchange. Calcium 215-222 hypocretin Mus musculus 13-17 14999052-8 2004 Nifedipine (10 microM), inhibited Hcrt/Orx responses but was more effective at abolishing spiking than plateau responses. Nifedipine 0-10 hypocretin Mus musculus 34-38 14999052-13 2004 Accordingly, the loss of Hcrt/Orx signaling in narcolepsy would be expected to disrupt calcium-dependent processes in these and other target structures. Calcium 87-94 hypocretin Mus musculus 25-29 12797956-3 2003 Activity of isolated orexin neurons is inhibited by glucose and leptin and stimulated by ghrelin. Glucose 52-59 hypocretin Mus musculus 21-27 12890892-2 2003 Forskolin-stimulated cyclic adenosine 3,5-monophosphate (cAMP) accumulation in OX2R-expressing cells was inhibited by orexin in a dose-dependent manner, and the effect was abolished by pretreatment with pertussis toxin (PTX). Colforsin 0-9 hypocretin Mus musculus 118-124 12890892-2 2003 Forskolin-stimulated cyclic adenosine 3,5-monophosphate (cAMP) accumulation in OX2R-expressing cells was inhibited by orexin in a dose-dependent manner, and the effect was abolished by pretreatment with pertussis toxin (PTX). Cyclic AMP 21-55 hypocretin Mus musculus 118-124 12890892-2 2003 Forskolin-stimulated cyclic adenosine 3,5-monophosphate (cAMP) accumulation in OX2R-expressing cells was inhibited by orexin in a dose-dependent manner, and the effect was abolished by pretreatment with pertussis toxin (PTX). Cyclic AMP 57-61 hypocretin Mus musculus 118-124 12890892-3 2003 The inhibitory effect of orexin on forskolin-stimulated cAMP accumulation was not observed in OX1R-expressing cells. Colforsin 35-44 hypocretin Mus musculus 25-31 12890892-3 2003 The inhibitory effect of orexin on forskolin-stimulated cAMP accumulation was not observed in OX1R-expressing cells. Cyclic AMP 56-60 hypocretin Mus musculus 25-31 12890892-4 2003 Administration of orexin to these cells resulted in a transient increase of intracellular calcium concentration ([Ca(2+)](i)). Calcium 90-97 hypocretin Mus musculus 18-24 12890892-8 2003 We found that a phospholipase C (PLC)-inhibitor, U73122, inhibits orexin-mediated neuronal activation, but PTX showed no effect on it. 1-(6-((3-methoxyestra-1,3,5(10)-trien-17-yl)amino)hexyl)-1H-pyrrole-2,5-dione 49-55 hypocretin Mus musculus 66-72 12797956-4 2003 Orexin expression of normal and ob/ob mice correlates negatively with changes in blood glucose, leptin, and food intake. Glucose 87-94 hypocretin Mus musculus 0-6 12560202-5 2003 Results obtained in obesity-prone rats and mice revealed a similar increase in orexin in close relation to triglycerides. Triglycerides 107-120 hypocretin Mus musculus 79-85 12495630-0 2002 Hypocretin/Orexin excites hypocretin neurons via a local glutamate neuron-A potential mechanism for orchestrating the hypothalamic arousal system. Glutamic Acid 57-66 hypocretin Mus musculus 0-17 12832517-5 2003 Acting on postsynaptic orexin type 2 receptors, orexin activates a sodium-calcium exchange current, thereby depolarizing the cell and increasing its firing frequency. Sodium 67-73 hypocretin Mus musculus 23-29 12832517-5 2003 Acting on postsynaptic orexin type 2 receptors, orexin activates a sodium-calcium exchange current, thereby depolarizing the cell and increasing its firing frequency. Sodium 67-73 hypocretin Mus musculus 48-54 12716916-0 2003 Involvement of the lateral hypothalamic peptide orexin in morphine dependence and withdrawal. Morphine 58-66 hypocretin Mus musculus 48-54 12716916-5 2003 In this study, we show that orexin neurons, and not nearby LH neurons expressing melanin-concentrating hormone (MCH), have mu-opioid receptors and respond to chronic morphine administration and opiate antagonist-precipitated morphine withdrawal. Morphine 166-174 hypocretin Mus musculus 28-34 12716916-5 2003 In this study, we show that orexin neurons, and not nearby LH neurons expressing melanin-concentrating hormone (MCH), have mu-opioid receptors and respond to chronic morphine administration and opiate antagonist-precipitated morphine withdrawal. Opiate Alkaloids 194-200 hypocretin Mus musculus 28-34 12716916-5 2003 In this study, we show that orexin neurons, and not nearby LH neurons expressing melanin-concentrating hormone (MCH), have mu-opioid receptors and respond to chronic morphine administration and opiate antagonist-precipitated morphine withdrawal. Morphine 225-233 hypocretin Mus musculus 28-34 12716916-6 2003 cAMP response element-mediated transcription is induced in a subset of orexin cells, but not MCH cells, after exposure to chronic morphine or induction of withdrawal. Cyclic AMP 0-4 hypocretin Mus musculus 71-77 12716916-6 2003 cAMP response element-mediated transcription is induced in a subset of orexin cells, but not MCH cells, after exposure to chronic morphine or induction of withdrawal. Morphine 130-138 hypocretin Mus musculus 71-77 12716916-7 2003 Additionally, c-Fos and the orexin gene itself are induced in orexin cells in the LH during morphine withdrawal. Morphine 92-100 hypocretin Mus musculus 28-34 12716916-7 2003 Additionally, c-Fos and the orexin gene itself are induced in orexin cells in the LH during morphine withdrawal. Morphine 92-100 hypocretin Mus musculus 62-68 12716916-8 2003 Finally, we show that orexin knock-out mice develop attenuated morphine dependence, as indicated by a less severe antagonist-precipitated withdrawal syndrome. Morphine 63-71 hypocretin Mus musculus 22-28 12716916-9 2003 Together, these studies support a role for the orexin system in molecular adaptations to morphine, and demonstrate dramatic differences in molecular responses among different populations of LH neurons. Morphine 89-97 hypocretin Mus musculus 47-53 12832517-5 2003 Acting on postsynaptic orexin type 2 receptors, orexin activates a sodium-calcium exchange current, thereby depolarizing the cell and increasing its firing frequency. Calcium 74-81 hypocretin Mus musculus 23-29 12832517-5 2003 Acting on postsynaptic orexin type 2 receptors, orexin activates a sodium-calcium exchange current, thereby depolarizing the cell and increasing its firing frequency. Calcium 74-81 hypocretin Mus musculus 48-54 12832517-6 2003 Because GABA is a potent stimulus for feeding, in both the ARC and its main projection site, these results suggest a mechanism for how orexin may control appetite. gamma-Aminobutyric Acid 8-12 hypocretin Mus musculus 135-141 12646175-5 2003 The neurotransmitters, which were implicated in sleep/wake regulation, affected the activity of orexin neurons; noradrenaline and serotonin hyperpolarized, while carbachol depolarized orexin neurons in either the presence or absence of tetrodotoxin. Norepinephrine 112-125 hypocretin Mus musculus 96-102 12646175-5 2003 The neurotransmitters, which were implicated in sleep/wake regulation, affected the activity of orexin neurons; noradrenaline and serotonin hyperpolarized, while carbachol depolarized orexin neurons in either the presence or absence of tetrodotoxin. Serotonin 130-139 hypocretin Mus musculus 96-102 12646175-5 2003 The neurotransmitters, which were implicated in sleep/wake regulation, affected the activity of orexin neurons; noradrenaline and serotonin hyperpolarized, while carbachol depolarized orexin neurons in either the presence or absence of tetrodotoxin. Carbachol 162-171 hypocretin Mus musculus 96-102 12646175-5 2003 The neurotransmitters, which were implicated in sleep/wake regulation, affected the activity of orexin neurons; noradrenaline and serotonin hyperpolarized, while carbachol depolarized orexin neurons in either the presence or absence of tetrodotoxin. Carbachol 162-171 hypocretin Mus musculus 184-190 12646175-5 2003 The neurotransmitters, which were implicated in sleep/wake regulation, affected the activity of orexin neurons; noradrenaline and serotonin hyperpolarized, while carbachol depolarized orexin neurons in either the presence or absence of tetrodotoxin. Tetrodotoxin 236-248 hypocretin Mus musculus 96-102 12495630-4 2002 The mechanism for this appears to be hypocretin-mediated excitation of local glutamatergic neurons that regulate hypocretin neuron activity, in part by presynaptic facilitation of glutamate release. Glutamic Acid 77-86 hypocretin Mus musculus 37-47 12495630-4 2002 The mechanism for this appears to be hypocretin-mediated excitation of local glutamatergic neurons that regulate hypocretin neuron activity, in part by presynaptic facilitation of glutamate release. Glutamic Acid 77-86 hypocretin Mus musculus 113-123 12388591-0 2002 Convergent excitation of dorsal raphe serotonin neurons by multiple arousal systems (orexin/hypocretin, histamine and noradrenaline). Serotonin 38-47 hypocretin Mus musculus 85-91 12388591-0 2002 Convergent excitation of dorsal raphe serotonin neurons by multiple arousal systems (orexin/hypocretin, histamine and noradrenaline). Serotonin 38-47 hypocretin Mus musculus 92-102 12217429-6 2002 Orexin-A at 3nmol improved retention in young and old SAMP8 mice. 3nmol 12-17 hypocretin Mus musculus 0-8 12388591-5 2002 In extracellular recordings, all three agonists increased the firing rate of serotonin neurons; the excitatory effects of histamine and orexin A were occluded by previous application of phenylephrine, suggesting that all three systems act via common effector mechanisms. Serotonin 77-86 hypocretin Mus musculus 136-144 12388591-5 2002 In extracellular recordings, all three agonists increased the firing rate of serotonin neurons; the excitatory effects of histamine and orexin A were occluded by previous application of phenylephrine, suggesting that all three systems act via common effector mechanisms. Phenylephrine 186-199 hypocretin Mus musculus 136-144 12388591-11 2002 Instead, in many cases the orexin A-induced current reversed in calcium-free medium at a value (-23 mV) consistent with the activation of a mixed cation channel (with relative permeabilities for sodium and potassium of 0.43 and 1, respectively). Calcium 64-71 hypocretin Mus musculus 27-35 12388591-11 2002 Instead, in many cases the orexin A-induced current reversed in calcium-free medium at a value (-23 mV) consistent with the activation of a mixed cation channel (with relative permeabilities for sodium and potassium of 0.43 and 1, respectively). Sodium 195-201 hypocretin Mus musculus 27-35 12388591-11 2002 Instead, in many cases the orexin A-induced current reversed in calcium-free medium at a value (-23 mV) consistent with the activation of a mixed cation channel (with relative permeabilities for sodium and potassium of 0.43 and 1, respectively). Potassium 206-215 hypocretin Mus musculus 27-35 12239605-3 2002 This study was undertaken to investigate the effects of AGRP, orexin and MCH on oxygen consumption. Oxygen 80-86 hypocretin Mus musculus 62-68 12239605-6 2002 Orexin (1 nmol/mouse) significantly increased oxygen consumption, while MCH (1 nmol/mouse) had no significant effect compared to ACSF-treated controls. Oxygen 46-52 hypocretin Mus musculus 0-6 11689192-5 2001 On the other hand, in the presence of atropine and guanethidine, orexin A induced a transient gradual relaxation in duodenal, jejunal and ileal segments. Atropine 38-46 hypocretin Mus musculus 65-73 12144948-6 2002 Intracerebroventricular administration of orexin-A and orexin-B was associated with glucose-lowering effects in fasting diabetic mice. Glucose 84-91 hypocretin Mus musculus 42-50 12144948-6 2002 Intracerebroventricular administration of orexin-A and orexin-B was associated with glucose-lowering effects in fasting diabetic mice. Glucose 84-91 hypocretin Mus musculus 55-63 11923451-6 2002 Postsynaptically, Hcrt/Orx produced an inward current and an increase in membrane current noise, which were accompanied by a conductance increase. orx 23-26 hypocretin Mus musculus 18-22 11689192-5 2001 On the other hand, in the presence of atropine and guanethidine, orexin A induced a transient gradual relaxation in duodenal, jejunal and ileal segments. Guanethidine 51-63 hypocretin Mus musculus 65-73 11689192-9 2001 The exogenous orexin A-induced relaxation was completely inhibited by L-NOARG. Nitroarginine 70-77 hypocretin Mus musculus 14-22 10481909-3 1999 Moreover, modafinil, an anti-narcoleptic drug with ill-defined mechanisms of action, activates orexin-containing neurons. Modafinil 10-19 hypocretin Mus musculus 95-101 11493714-5 2001 Microdialysis studies showed that application of orexin A to the TMN increased histamine release from both the medial preoptic area and the frontal cortex by approximately 2-fold over the baseline for 80 to 160 min in a dose-dependent manner. Histamine 79-88 hypocretin Mus musculus 49-57 11394998-3 2001 We generated transgenic mice in which orexin-containing neurons are ablated by orexinergic-specific expression of a truncated Machado-Joseph disease gene product (ataxin-3) with an expanded polyglutamine stretch. polyglutamine 190-203 hypocretin Mus musculus 38-44 10997654-16 2000 The LHA is important in receiving sensory signals from the gut and liver, and in sensing glucose, and orexin neurones may be involved in stimulating feeding in response to falls in plasma glucose. Glucose 188-195 hypocretin Mus musculus 102-108 10191330-5 1999 Double-labeling experiments in mice combining retrograde transport of diamidino yellow after spinal cord injections and immunocytochemistry support the concept that hypocretin-immunoreactive fibers in the cord originate from the neurons in the lateral hypothalamus. diamidino yellow 70-86 hypocretin Mus musculus 165-175 10191330-6 1999 Digital-imaging physiological studies with fura-2 detected a rise in intracellular calcium in response to hypocretin in cultured rat spinal cord neurons, indicating that spinal cord neurons express hypocretin-responsive receptors. Fura-2 43-49 hypocretin Mus musculus 106-116 10191330-6 1999 Digital-imaging physiological studies with fura-2 detected a rise in intracellular calcium in response to hypocretin in cultured rat spinal cord neurons, indicating that spinal cord neurons express hypocretin-responsive receptors. Fura-2 43-49 hypocretin Mus musculus 198-208 10191330-6 1999 Digital-imaging physiological studies with fura-2 detected a rise in intracellular calcium in response to hypocretin in cultured rat spinal cord neurons, indicating that spinal cord neurons express hypocretin-responsive receptors. Calcium 83-90 hypocretin Mus musculus 106-116 10191330-6 1999 Digital-imaging physiological studies with fura-2 detected a rise in intracellular calcium in response to hypocretin in cultured rat spinal cord neurons, indicating that spinal cord neurons express hypocretin-responsive receptors. Calcium 83-90 hypocretin Mus musculus 198-208 34992518-11 2021 Physical or molecular interactions between these two systems may provide valuable insight into drug-drug interactions between cannabinoid and orexin drugs for the treatment of insomnia, pain, and other disorders. Cannabinoids 126-137 hypocretin Mus musculus 142-148 10087007-9 1999 The octanol/buffer partition coefficient of 0.232 showed that orexin A was highly lipophilic, whereas the value for orexin B was only 0.030. Octanols 4-11 hypocretin Mus musculus 62-70 34853083-0 2022 Hypocretin/orexin interactions with norepinephrine contribute to the opiate withdrawal syndrome. Norepinephrine 36-50 hypocretin Mus musculus 0-10 34853083-0 2022 Hypocretin/orexin interactions with norepinephrine contribute to the opiate withdrawal syndrome. Norepinephrine 36-50 hypocretin Mus musculus 11-17 34853083-1 2022 We previously found that human heroin addicts and mice chronically exposed to morphine exhibit a significant increase in the number of detected hypocretin/orexin (Hcrt) producing neurons. Heroin 31-37 hypocretin Mus musculus 144-161 34853083-1 2022 We previously found that human heroin addicts and mice chronically exposed to morphine exhibit a significant increase in the number of detected hypocretin/orexin (Hcrt) producing neurons. Heroin 31-37 hypocretin Mus musculus 163-167 34853083-1 2022 We previously found that human heroin addicts and mice chronically exposed to morphine exhibit a significant increase in the number of detected hypocretin/orexin (Hcrt) producing neurons. Morphine 78-86 hypocretin Mus musculus 144-161 34853083-1 2022 We previously found that human heroin addicts and mice chronically exposed to morphine exhibit a significant increase in the number of detected hypocretin/orexin (Hcrt) producing neurons. Morphine 78-86 hypocretin Mus musculus 163-167 34666119-0 2021 Orexin-B exerts excitatory effects on nigral dopaminergic neurons and alleviates motor disorders in MPTP parkinsonian mice. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 100-104 hypocretin Mus musculus 0-8 34666119-2 2021 The present study showed that orexin-B exerted marked excitatory effects via orexin-2 receptor on the nigral dopaminergic neurons in MPTP parkinsonian mice, while blocking orexin-2 receptor decreased the firing rate of dopaminergic neurons significantly. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 133-137 hypocretin Mus musculus 30-38 34666119-3 2021 Furthermore, intracerebroventricular application of orexin-B relieved the degeneration of dopaminergic neurons, increased the general spontaneous activity and alleviated motor coordination in MPTP parkinsonian mice. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 192-196 hypocretin Mus musculus 52-60 34101446-0 2021 Discovery of Arylsulfonamides as Dual Orexin Receptor Agonists. arylsulfonamides 13-29 hypocretin Mus musculus 38-44 34702886-6 2021 In WT mice, DOCA-salt-treatment increased gene and protein expression of orexin A, orexin receptor 1, and orexin receptor 2 in the hypothalamic paraventricular nucleus and these effects were attenuated in B1RKO mice. doca-salt 12-21 hypocretin Mus musculus 73-81 34475397-6 2021 Optogenetic activation of orexin neurons in lateral hypothalamus or orexinergic fibers innervating raphe pallidus impaired or improved glucose tolerance, respectively. Glucose 135-142 hypocretin Mus musculus 26-32 34458633-10 2021 In contrast, neuronal populations expressing orexin, oxytocin, vasopressin as well as tyrosine hydroxylase in the A13 area are resistant to QA-induced toxicity. Quinolinic Acid 140-142 hypocretin Mus musculus 45-51 34093114-8 2021 These data support the view that orexin peptide deficiency is sufficient to increase histamine neuron number, supporting the involvement of the histamine waking system in the pathophysiology of NT1. Histamine 85-94 hypocretin Mus musculus 33-39 34225941-8 2021 We found that in mice, chronic morphine administration induced the same changes in Hcrt neuron number and size. Morphine 31-39 hypocretin Mus musculus 83-87 34225941-13 2021 These findings are consistent with the concept that an increased number of Hcrt neurons may underlie and maintain opioid or cocaine use disorders. Cocaine 124-131 hypocretin Mus musculus 75-79 35101602-0 2022 Orexin deficiency affects sensorimotor gating and its amphetamine-induced impairment. Amphetamine 54-65 hypocretin Mus musculus 0-6 35101602-3 2022 Utilizing orexin-deficient mice, the present study tested the hypothesis that orexin is involved in two further mouse behavioral endophenotypes of neuropsychiatric disorders, i.e., sensorimotor gating and amphetamine sensitivity. Amphetamine 205-216 hypocretin Mus musculus 78-84 35101602-5 2022 Amphetamine treatment impaired prepulse inhibition in wildtype and heterozygous orexin-deficient mice, but had no effects in homozygous orexin-deficient mice. Amphetamine 0-11 hypocretin Mus musculus 80-86 35101602-7 2022 These data indicate that the orexin system modulates prepulse inhibition and is involved in mediating amphetamine"s effect on prepulse inhibition. Amphetamine 102-113 hypocretin Mus musculus 29-35 34093114-4 2021 The aim of this study was to test whether histamine neurons are increased in number in orexin-deficient mice and whether orexin neurons are decreased in number in histamine-deficient mice. Histamine 163-172 hypocretin Mus musculus 121-127 35115701-4 2022 Further, systemic IGF-I modified the GABA/Glutamate synaptic structure in orexin neurons of naive wild-type mice by increasing the dephosphorylation of GABA(B) receptor subunit through inhibition of AMP-kinase (AMPK). gamma-Aminobutyric Acid 152-156 hypocretin Mus musculus 74-80 35477218-4 2022 Here we investigated the role of the endogenous cannabinoid system in the impaired fear extinction induced by orexin-A (OXA) in male mice. Cannabinoids 48-59 hypocretin Mus musculus 110-118 33078457-0 2021 Stress induces reinstatement of extinguished cocaine conditioned place preference by a sequential signaling via neuropeptide S, orexin, and endocannabinoid. Cocaine 45-52 hypocretin Mus musculus 128-134 35201886-6 2022 Disruption of Kcnq2/3 genes in Hcrt neurons of young mice destabilized sleep, mimicking aging-associated sleep fragmentation, whereas the KCNQ-selective activator flupirtine hyperpolarized Hcrt neurons and rejuvenated sleep architecture in aged mice. flupirtine 163-173 hypocretin Mus musculus 189-193 35182424-4 2022 We compared the development of narcoleptic symptomatology in male vs. female orexin-tTA; TetO-DTA mice, a model in which Hcrt neuron degeneration can be initiated by removal of doxycycline (DOX) from the diet. Doxycycline 177-188 hypocretin Mus musculus 121-125 35182424-4 2022 We compared the development of narcoleptic symptomatology in male vs. female orexin-tTA; TetO-DTA mice, a model in which Hcrt neuron degeneration can be initiated by removal of doxycycline (DOX) from the diet. Doxycycline 190-193 hypocretin Mus musculus 121-125 33550481-12 2021 Modulation in cortical levels of histamine, norepinephrine and dopamine provides the neurochemical basis for wake-promoting and anticataplectic effects observed in orexin knockout mice. Histamine 33-42 hypocretin Mus musculus 164-170 33550481-12 2021 Modulation in cortical levels of histamine, norepinephrine and dopamine provides the neurochemical basis for wake-promoting and anticataplectic effects observed in orexin knockout mice. Norepinephrine 44-58 hypocretin Mus musculus 164-170 33550481-12 2021 Modulation in cortical levels of histamine, norepinephrine and dopamine provides the neurochemical basis for wake-promoting and anticataplectic effects observed in orexin knockout mice. Dopamine 63-71 hypocretin Mus musculus 164-170 34051297-9 2021 Behavioral tests revealed that CIH significantly increased the escape latency and time of arriving platform, of which were markedly decreased by OXA treatment. cih 31-34 hypocretin Mus musculus 145-148 34051297-12 2021 Furthermore, we found that micro-injection of OXA attenuated CIH-induced apoptotic cell death and oxidative stress in the hippocampus. cih 61-64 hypocretin Mus musculus 46-49 34051297-13 2021 Our results suggested that OXA might improve cognitive impairment induced by CIH through inhibiting hippocampal apoptosis and oxidative stress. cih 77-80 hypocretin Mus musculus 27-30 35246205-5 2022 We first prepared orexin-knockout mice crossed with transgenic mice carrying a tetracycline-controlled transactivator transgene under the control of the orexin promoter. Tetracycline 79-91 hypocretin Mus musculus 153-159 35154573-0 2022 Electroacupuncture Enhances Neuroplasticity by Regulating the Orexin A-Mediated cAMP/PKA/CREB Signaling Pathway in Senescence-Accelerated Mouse Prone 8 (SAMP8) Mice. Cyclic AMP 80-84 hypocretin Mus musculus 62-70 35128515-0 2022 Multifaceted roles of orexin neurons in mediating methamphetamine-induced changes in body temperature and heart rate. Methamphetamine 50-65 hypocretin Mus musculus 22-28 35128515-11 2022 These results suggest that glutamate and orexin from orexin neurons have differential roles in mediating METH-induced changes in body temperature and heart rate. Methamphetamine 105-109 hypocretin Mus musculus 41-47 33078457-2 2021 Previously, we reported that orexins released during stress, via orexin OX1 receptors (OX1 Rs), contribute to the reinstatement of cocaine seeking through endocannabinoid/CB1 receptor (CB1 R)-mediated dopaminergic disinhibition in the ventral tegmental area (VTA). Cocaine 131-138 hypocretin Mus musculus 29-35 33078457-3 2021 Here, we further demonstrated that NPS released during stress is an up-stream activator of this orexin-endocannabinoid cascade in the VTA, leading to the reinstatement of cocaine seeking. Endocannabinoids 103-118 hypocretin Mus musculus 96-102 33078457-3 2021 Here, we further demonstrated that NPS released during stress is an up-stream activator of this orexin-endocannabinoid cascade in the VTA, leading to the reinstatement of cocaine seeking. Cocaine 171-178 hypocretin Mus musculus 96-102 33069869-13 2021 PERSPECTIVES: Median nerve stimulation relieves neuropathic pain in mice without tolerance and retains efficacy even in mice with analgesic tolerance to escalating doses of morphine, via an opioid-independent, orexin-endocannabinoid-mediated mechanism. Morphine 173-181 hypocretin Mus musculus 210-216 33746708-5 2021 When doxycycline is removed from the diet, the orexin neurons of these mice express diphtheria toxin A and die within 2-3 weeks. Doxycycline 5-16 hypocretin Mus musculus 47-53 32653557-0 2021 Enhancement of the rewarding effects of 3,4-methylenedioxymethamphetamine in orexin knockout mice. N-Methyl-3,4-methylenedioxyamphetamine 40-73 hypocretin Mus musculus 77-83 33613258-0 2021 Orexin-A/Hypocretin-1 Controls the VTA-NAc Mesolimbic Pathway via Endocannabinoid-Mediated Disinhibition of Dopaminergic Neurons in Obese Mice. nac 39-42 hypocretin Mus musculus 0-8 33613258-0 2021 Orexin-A/Hypocretin-1 Controls the VTA-NAc Mesolimbic Pathway via Endocannabinoid-Mediated Disinhibition of Dopaminergic Neurons in Obese Mice. Endocannabinoids 66-81 hypocretin Mus musculus 0-8 32653557-7 2021 Interestingly, we found that MDMA induced a conditioned place preference in orexin KO mice, but not in wild type (WT) mice. N-Methyl-3,4-methylenedioxyamphetamine 29-33 hypocretin Mus musculus 76-82 32653557-8 2021 In addition, MDMA produced methylphenidate/methamphetamine-like discriminative stimulus effects in orexin KO mice, but not WT mice. N-Methyl-3,4-methylenedioxyamphetamine 13-17 hypocretin Mus musculus 99-105 32653557-8 2021 In addition, MDMA produced methylphenidate/methamphetamine-like discriminative stimulus effects in orexin KO mice, but not WT mice. Methylphenidate 27-42 hypocretin Mus musculus 99-105 32653557-8 2021 In addition, MDMA produced methylphenidate/methamphetamine-like discriminative stimulus effects in orexin KO mice, but not WT mice. Methamphetamine 43-58 hypocretin Mus musculus 99-105 32653557-10 2021 In substitution tests using a drug discrimination procedure, substitution of 5-HT1A receptor agonist for the discriminative stimulus effects of methylphenidate was enhanced in orexin KO mice. Methylphenidate 144-159 hypocretin Mus musculus 176-182 32653557-13 2021 On the other hand, deficiencies in orexin may enhance the abuse liability of MDMA by changing a postsynaptic signal transduction accompanied by changes in discriminative stimulus effects themselves. N-Methyl-3,4-methylenedioxyamphetamine 77-81 hypocretin Mus musculus 35-41 33139319-4 2020 Exposure to orexin-receptor antagonism (1 mg/kg SB334867, an orexin 1 receptor antagonist) just prior to cocaine-conditioning trials or the post-conditioning test, attenuated SD-enhanced preference. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 48-56 hypocretin Mus musculus 12-18 33139319-4 2020 Exposure to orexin-receptor antagonism (1 mg/kg SB334867, an orexin 1 receptor antagonist) just prior to cocaine-conditioning trials or the post-conditioning test, attenuated SD-enhanced preference. Cocaine 105-112 hypocretin Mus musculus 12-18 33139319-5 2020 This suggests a potential therapeutic role for the manipulation of the orexin system to mitigate drug seeking, especially in the context of sleep loss prior to drug exposure.Significance statement Drugs of abuse, including cocaine, disturb sleep and sleep disturbance has been implicated in probability of relapse; however, there have been few direct tests of sleep disturbance on drug seeking behavior. Cocaine 223-230 hypocretin Mus musculus 71-77 33139319-7 2020 Furthermore, antagonism of the orexin system, a neuromodulator involved in motivation-based arousal, reduces this SD-induced enhancement of cocaine preference. Cocaine 140-147 hypocretin Mus musculus 31-37 32667686-8 2020 Correspondingly, calcium recordings revealed that orexin cell activity rapidly reduced upon exiting the innately-aversive places. Calcium 17-24 hypocretin Mus musculus 50-56 32084419-3 2020 In this study, we tested the role of orexin-A in CPSP induction in mice. cpsp 49-53 hypocretin Mus musculus 37-45 32575773-0 2020 Role of 2-Arachidonoyl-Glycerol and CB1 Receptors in Orexin-A-Mediated Prevention of Oxygen-Glucose Deprivation-Induced Neuronal Injury. glyceryl 2-arachidonate 8-31 hypocretin Mus musculus 53-61 32575773-2 2020 Since the endocannabinoid 2-arachidonoylglycerol (2-AG) and cannabinoid type-1 (CB1) receptors were previously shown to mediate some of the effects of OX-A exerted through the orexin-1 receptor (OX-1R), we investigated the involvement of 2-AG in OX-A-induced neuroprotection following oxygen and glucose deprivation (OGD) in mouse cortical neurons. glyceryl 2-arachidonate 26-48 hypocretin Mus musculus 151-155 32575773-2 2020 Since the endocannabinoid 2-arachidonoylglycerol (2-AG) and cannabinoid type-1 (CB1) receptors were previously shown to mediate some of the effects of OX-A exerted through the orexin-1 receptor (OX-1R), we investigated the involvement of 2-AG in OX-A-induced neuroprotection following oxygen and glucose deprivation (OGD) in mouse cortical neurons. glyceryl 2-arachidonate 26-48 hypocretin Mus musculus 246-250 32575773-2 2020 Since the endocannabinoid 2-arachidonoylglycerol (2-AG) and cannabinoid type-1 (CB1) receptors were previously shown to mediate some of the effects of OX-A exerted through the orexin-1 receptor (OX-1R), we investigated the involvement of 2-AG in OX-A-induced neuroprotection following oxygen and glucose deprivation (OGD) in mouse cortical neurons. glyceryl 2-arachidonate 50-54 hypocretin Mus musculus 151-155 32575773-2 2020 Since the endocannabinoid 2-arachidonoylglycerol (2-AG) and cannabinoid type-1 (CB1) receptors were previously shown to mediate some of the effects of OX-A exerted through the orexin-1 receptor (OX-1R), we investigated the involvement of 2-AG in OX-A-induced neuroprotection following oxygen and glucose deprivation (OGD) in mouse cortical neurons. glyceryl 2-arachidonate 50-54 hypocretin Mus musculus 246-250 32575773-2 2020 Since the endocannabinoid 2-arachidonoylglycerol (2-AG) and cannabinoid type-1 (CB1) receptors were previously shown to mediate some of the effects of OX-A exerted through the orexin-1 receptor (OX-1R), we investigated the involvement of 2-AG in OX-A-induced neuroprotection following oxygen and glucose deprivation (OGD) in mouse cortical neurons. glyceryl 2-arachidonate 238-242 hypocretin Mus musculus 151-155 32575773-2 2020 Since the endocannabinoid 2-arachidonoylglycerol (2-AG) and cannabinoid type-1 (CB1) receptors were previously shown to mediate some of the effects of OX-A exerted through the orexin-1 receptor (OX-1R), we investigated the involvement of 2-AG in OX-A-induced neuroprotection following oxygen and glucose deprivation (OGD) in mouse cortical neurons. Oxygen 285-291 hypocretin Mus musculus 151-155 32575773-3 2020 OGD-induced reactive oxygen species (ROS) accumulation and neuronal death were prevented by both OX-A and arachidonyl-2"-chloroethylamide (ACEA), a synthetic CB1 receptor agonist, in a manner sensitive to OX-1R and CB1 receptor antagonists, SB334867 and AM251. Reactive Oxygen Species 12-35 hypocretin Mus musculus 97-101 32575773-3 2020 OGD-induced reactive oxygen species (ROS) accumulation and neuronal death were prevented by both OX-A and arachidonyl-2"-chloroethylamide (ACEA), a synthetic CB1 receptor agonist, in a manner sensitive to OX-1R and CB1 receptor antagonists, SB334867 and AM251. Reactive Oxygen Species 37-40 hypocretin Mus musculus 97-101 32575773-3 2020 OGD-induced reactive oxygen species (ROS) accumulation and neuronal death were prevented by both OX-A and arachidonyl-2"-chloroethylamide (ACEA), a synthetic CB1 receptor agonist, in a manner sensitive to OX-1R and CB1 receptor antagonists, SB334867 and AM251. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 241-249 hypocretin Mus musculus 97-101 32575773-3 2020 OGD-induced reactive oxygen species (ROS) accumulation and neuronal death were prevented by both OX-A and arachidonyl-2"-chloroethylamide (ACEA), a synthetic CB1 receptor agonist, in a manner sensitive to OX-1R and CB1 receptor antagonists, SB334867 and AM251. AM 251 254-259 hypocretin Mus musculus 97-101 32575773-4 2020 OX-A stimulated 2-AG biosynthesis in cortical neurons. glyceryl 2-arachidonate 16-20 hypocretin Mus musculus 0-4 32575773-6 2020 OX-A-induced neuroprotection was mediated by the phosphoinositide-3-kinase/Akt (PI3K/Akt) survival pathway since both OX-A and ACEA induced phosphorylation of Akt and prevented OGD-induced cytochrome c release from the mitochondria, in a manner counteracted by SB334867 or AM251. arachidonyl-2-chloroethylamide 127-131 hypocretin Mus musculus 0-4 32575773-6 2020 OX-A-induced neuroprotection was mediated by the phosphoinositide-3-kinase/Akt (PI3K/Akt) survival pathway since both OX-A and ACEA induced phosphorylation of Akt and prevented OGD-induced cytochrome c release from the mitochondria, in a manner counteracted by SB334867 or AM251. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 261-269 hypocretin Mus musculus 0-4 32575773-6 2020 OX-A-induced neuroprotection was mediated by the phosphoinositide-3-kinase/Akt (PI3K/Akt) survival pathway since both OX-A and ACEA induced phosphorylation of Akt and prevented OGD-induced cytochrome c release from the mitochondria, in a manner counteracted by SB334867 or AM251. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 261-269 hypocretin Mus musculus 118-122 32575773-6 2020 OX-A-induced neuroprotection was mediated by the phosphoinositide-3-kinase/Akt (PI3K/Akt) survival pathway since both OX-A and ACEA induced phosphorylation of Akt and prevented OGD-induced cytochrome c release from the mitochondria, in a manner counteracted by SB334867 or AM251. AM 251 273-278 hypocretin Mus musculus 0-4 32575773-6 2020 OX-A-induced neuroprotection was mediated by the phosphoinositide-3-kinase/Akt (PI3K/Akt) survival pathway since both OX-A and ACEA induced phosphorylation of Akt and prevented OGD-induced cytochrome c release from the mitochondria, in a manner counteracted by SB334867 or AM251. AM 251 273-278 hypocretin Mus musculus 118-122 32575773-7 2020 Administration of OX-A reduced infarct volume and elevated brain 2-AG levels in a mouse model of transient ischemia. glyceryl 2-arachidonate 65-69 hypocretin Mus musculus 18-22 32575773-8 2020 These results suggest that 2-AG and CB1 receptor mediate OX-A prevention of ischemia-induced neuronal apoptosis. glyceryl 2-arachidonate 27-31 hypocretin Mus musculus 57-61 31830270-8 2020 Collectively, these results demonstrate that DORA"s potently increase NREM and REM sleep in mice via blockade of orexin signaling, and higher doses can cause cataplexy when co-administered with a likely rewarding stimulus. dual orexin receptor antagonist 12 45-49 hypocretin Mus musculus 113-119 33224245-0 2020 Suvorexant, a Dual Orexin Receptor Antagonist, Protected Seizure through Interaction with GABAA and Glutamate Receptors. suvorexant 0-10 hypocretin Mus musculus 19-25 33224245-2 2020 In this study, the effects of suvorexant (orexin receptor antagonist) on pentylenetetrazol (PTZ) and maximal electroshock (MES)-induced seizure were investigated. Pentylenetetrazole 73-90 hypocretin Mus musculus 42-48 32030748-12 2020 Blocking orexin receptors prevented the methacholine (MCh)-induced increase in breathing frequency in KO mice and reduced MCh-induced seizures, via a direct or indirect mechanism. Methacholine Chloride 40-52 hypocretin Mus musculus 9-15 32030748-12 2020 Blocking orexin receptors prevented the methacholine (MCh)-induced increase in breathing frequency in KO mice and reduced MCh-induced seizures, via a direct or indirect mechanism. Methacholine Chloride 54-57 hypocretin Mus musculus 9-15 32030748-12 2020 Blocking orexin receptors prevented the methacholine (MCh)-induced increase in breathing frequency in KO mice and reduced MCh-induced seizures, via a direct or indirect mechanism. Methacholine Chloride 122-125 hypocretin Mus musculus 9-15 32084419-8 2020 ICV injection of orexin-A dose-dependently suppressed BCAO-induced mechanical allodynia. bcao 54-58 hypocretin Mus musculus 17-25 31605778-6 2020 Both the sleep abnormalities and increased orexin expression in Tsc1GFAPCKO mice were reversed by rapamycin treatment, indicating their dependence on mTOR activation. Sirolimus 98-107 hypocretin Mus musculus 43-49 32118032-11 2020 The percentages of double labeled neurons in PFA and DMH orexin fields are significantly higher than those neurons in the LH of HbSS-BERK mice (* p < 0.05). 1,2-Dimethylhydrazine 53-56 hypocretin Mus musculus 57-63 31927055-8 2020 Using thioflavine-S staining and ELISA, we found that Orexin-A promoted Abeta accumulation in APP/PS1 mice. thioflavin T 6-17 hypocretin Mus musculus 54-62 31927055-9 2020 By evaluating mitochondrial morphology, cytochrome c oxidase activity, ATP level, mitochondrial DNA copy number, and reactive oxygen species, we found that Orexin-A aggravated mitochondrial impairment in APP/PS1 mice and Abeta-treated SH-SY5Y cells. Adenosine Triphosphate 71-74 hypocretin Mus musculus 156-164 31927055-9 2020 By evaluating mitochondrial morphology, cytochrome c oxidase activity, ATP level, mitochondrial DNA copy number, and reactive oxygen species, we found that Orexin-A aggravated mitochondrial impairment in APP/PS1 mice and Abeta-treated SH-SY5Y cells. Oxygen 126-132 hypocretin Mus musculus 156-164 31365289-3 2019 Here we showed that bilateral microinjection of both orexin-A and orexin-B into the globus pallidus alleviated motor deficits in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced parkinsonian mice. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 129-173 hypocretin Mus musculus 53-61 31724150-0 2020 Reduced c-Fos expression in orexin neurons of the lateral hypothalamic area and the locus coeruleus following injection of spinosin into mice. spinosin 123-131 hypocretin Mus musculus 28-34 31724150-6 2020 Compared to the saline injection, the application of spinosin also resulted in a marked decrease in c-Fos expression in the LHA orexin neurons. spinosin 53-61 hypocretin Mus musculus 128-134 31724150-7 2020 CONCLUSIONS: These findings suggest that spinosin administration results in a restricted pattern of c-Fos expression within the LHA orexin neurons and the LC, suggesting that this particular neuronal inactivation contributes to sedation and hypnosis. spinosin 41-49 hypocretin Mus musculus 132-138 30276922-4 2019 Both 10 and 20 mg/kg of cocaine induced similar magnitudes of CPP in mice and re-potentiation in their hippocampal slices, but differed in their susceptibility to TCS1102, a dual (OX1 and OX2 ) orexin receptor antagonist. Cocaine 24-31 hypocretin Mus musculus 194-200 30276922-9 2019 Conversely, SCH23390 prevented orexin A-induced hippocampal re-potentiation. SCH 23390 12-20 hypocretin Mus musculus 31-39 31667577-8 2019 In addition to the pathway via extracellular signal-regulated kinases, OXA triggered adenylyl cyclase-induced cAMP elevation, which results in protein kinase A-mediated activation of cAMP response element-binding proteins (CREB). Cyclic AMP 110-114 hypocretin Mus musculus 71-74 31667577-8 2019 In addition to the pathway via extracellular signal-regulated kinases, OXA triggered adenylyl cyclase-induced cAMP elevation, which results in protein kinase A-mediated activation of cAMP response element-binding proteins (CREB). Cyclic AMP 183-187 hypocretin Mus musculus 71-74 31365289-3 2019 Here we showed that bilateral microinjection of both orexin-A and orexin-B into the globus pallidus alleviated motor deficits in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced parkinsonian mice. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 129-173 hypocretin Mus musculus 66-74 31365289-3 2019 Here we showed that bilateral microinjection of both orexin-A and orexin-B into the globus pallidus alleviated motor deficits in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced parkinsonian mice. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 175-179 hypocretin Mus musculus 53-61 31365289-3 2019 Here we showed that bilateral microinjection of both orexin-A and orexin-B into the globus pallidus alleviated motor deficits in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced parkinsonian mice. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 175-179 hypocretin Mus musculus 66-74 31365289-7 2019 Orexin-A-induced increase in firing rate of pallidal neurons in MPTP parkinsonian mice was stronger than that of normal mice. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 64-68 hypocretin Mus musculus 0-8 30253175-2 2019 Cocaine exposure is associated with presynaptic plasticity at LHA orexin cells such that excitatory input to orexin cells is enhanced acutely and into withdrawal. Cocaine 0-7 hypocretin Mus musculus 66-72 31375626-8 2019 The transcriptome results were used to develop protocols for the production of HCRT and MCH neurons from induced pluripotent stem cells and ascorbic acid was found necessary for HCRT and BMP7 for MCH cell differentiation. Ascorbic Acid 140-153 hypocretin Mus musculus 178-182 30253175-2 2019 Cocaine exposure is associated with presynaptic plasticity at LHA orexin cells such that excitatory input to orexin cells is enhanced acutely and into withdrawal. Cocaine 0-7 hypocretin Mus musculus 109-115 30253175-4 2019 Studies in hypothalamic slices from drug-naive animals indicate that agonism of group III metabotropic glutamate receptors (mGluRs) reduces presynaptic glutamate release onto orexin cells. Glutamic Acid 103-112 hypocretin Mus musculus 175-181 30253175-6 2019 First, we verified that group III mGluRs regulate orexin cell activity in behaving animals by showing that intra-LHA infusions of the selective agonist L-(+)-2-Amino-4-phosphonobutyric acid (L-AP4) reduces c-fos expression in orexin cells following 24 h food deprivation. 2-amino-4-phosphono-propinate 152-189 hypocretin Mus musculus 50-56 30253175-6 2019 First, we verified that group III mGluRs regulate orexin cell activity in behaving animals by showing that intra-LHA infusions of the selective agonist L-(+)-2-Amino-4-phosphonobutyric acid (L-AP4) reduces c-fos expression in orexin cells following 24 h food deprivation. 2-amino-4-phosphono-propinate 152-189 hypocretin Mus musculus 226-232 30253175-11 2019 These effects are likely mediated, at least in part, by normalization of presynaptic plasticity at orexin cells that occurs as a result of cocaine exposure. Cocaine 139-146 hypocretin Mus musculus 99-105 30919010-5 2019 Cisplatin decreased the expression of prepro-orexin mRNA, which encodes precursors of orexin, in the hypothalamus of mice. Cisplatin 0-9 hypocretin Mus musculus 38-51 30919010-5 2019 Cisplatin decreased the expression of prepro-orexin mRNA, which encodes precursors of orexin, in the hypothalamus of mice. Cisplatin 0-9 hypocretin Mus musculus 45-51 30919010-7 2019 The effect of the H4 receptor antagonist on cisplatin-induced anorexia in mice was antagonized by an orexin OX2 receptor antagonist (JNJ10397049, 5 mg/kg) rather than an orexin OX1 receptor antagonist (SB408124, 30 mg/kg). Cisplatin 44-53 hypocretin Mus musculus 101-107 29462275-5 2019 Bath application of HCRT1/orexin-A evoked an inward current and membrane depolarization in most nNOS/NK1R cells which persisted in tetrodotoxin; optogenetic stimulation of Hcrt terminals expressing channelrhodopsin-2 confirmed these results, and pharmacological studies determined that HCRTR1 mediated these responses. Tetrodotoxin 131-143 hypocretin Mus musculus 26-34 31394498-0 2019 Chronotherapeutic effect of orexin antagonists on glucose metabolism in diabetic mice. Glucose 50-57 hypocretin Mus musculus 28-34 31159922-3 2019 However, orexin neurons produce and release orexin as well as several co-transmitters including dynorphin and glutamate. Glutamic Acid 110-119 hypocretin Mus musculus 9-15 31159923-7 2019 Moreover, VTAGad67+ directly innervated and inhibited wake-promoting orexin/hypocretin neurons by releasing GABA. gamma-Aminobutyric Acid 108-112 hypocretin Mus musculus 69-75 31159923-7 2019 Moreover, VTAGad67+ directly innervated and inhibited wake-promoting orexin/hypocretin neurons by releasing GABA. gamma-Aminobutyric Acid 108-112 hypocretin Mus musculus 76-86 31394498-3 2019 Resting-phase administration of dual orexin receptor antagonist (DORA) has been shown to improve sleep abnormalities and glucose intolerance in type 2 diabetic db/db mice, although the mechanism remains unknown. Glucose 121-128 hypocretin Mus musculus 37-43 30465812-9 2019 Finally, in vivo microdialysis experiments showed that amphetamine potentiated orexin A-induced dopamine and glutamate release in the ventral tegmental area (VTA). Amphetamine 55-66 hypocretin Mus musculus 79-87 30465812-9 2019 Finally, in vivo microdialysis experiments showed that amphetamine potentiated orexin A-induced dopamine and glutamate release in the ventral tegmental area (VTA). Dopamine 96-104 hypocretin Mus musculus 79-87 30465812-9 2019 Finally, in vivo microdialysis experiments showed that amphetamine potentiated orexin A-induced dopamine and glutamate release in the ventral tegmental area (VTA). Glutamic Acid 109-118 hypocretin Mus musculus 79-87 31029425-6 2019 Icv injection of an orexin receptor antagonist, SB334867 reduced STAT3 phosphorylation, which was enhanced by icv injection of orexin but not by direct injection of orexin into MBH. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 48-56 hypocretin Mus musculus 20-26 31029425-6 2019 Icv injection of an orexin receptor antagonist, SB334867 reduced STAT3 phosphorylation, which was enhanced by icv injection of orexin but not by direct injection of orexin into MBH. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 48-56 hypocretin Mus musculus 127-133 31029425-6 2019 Icv injection of an orexin receptor antagonist, SB334867 reduced STAT3 phosphorylation, which was enhanced by icv injection of orexin but not by direct injection of orexin into MBH. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 48-56 hypocretin Mus musculus 127-133 29462275-5 2019 Bath application of HCRT1/orexin-A evoked an inward current and membrane depolarization in most nNOS/NK1R cells which persisted in tetrodotoxin; optogenetic stimulation of Hcrt terminals expressing channelrhodopsin-2 confirmed these results, and pharmacological studies determined that HCRTR1 mediated these responses. Tetrodotoxin 131-143 hypocretin Mus musculus 172-176 30399595-3 2019 administration of orexin 1 (OX1) receptor antagonist -SB334867- alone or in combination with NO agents on depression using the forced swimming test (FST), tail suspension test (TST) and the number of crossings in open-field test (OFT) in mice. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 54-62 hypocretin Mus musculus 18-24 30365112-5 2019 In addition, reverse transcription-quantitative polymerase chain reaction and western blot analysis revealed that the expression levels of tau, p-tau, orexin A and orexin neurons express adenosine A1 receptor (A1R) were markedly upregulated in the brain tissue of AD mice compared with that in samples obtained from control mice. Adenosine 187-196 hypocretin Mus musculus 151-159 30365112-5 2019 In addition, reverse transcription-quantitative polymerase chain reaction and western blot analysis revealed that the expression levels of tau, p-tau, orexin A and orexin neurons express adenosine A1 receptor (A1R) were markedly upregulated in the brain tissue of AD mice compared with that in samples obtained from control mice. Adenosine 187-196 hypocretin Mus musculus 151-157 30905886-3 2019 The orexin receptor antagonist suvorexant, which specifically block the endogenous waking system, has been approved as a new drug to treat insomnia. suvorexant 31-41 hypocretin Mus musculus 4-10 29964087-5 2018 Binding of OXA to OX1R was blocked using an OX1R antagonist, SB-334867. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 61-70 hypocretin Mus musculus 11-14 30555297-2 2018 Orexin neurons send excitatory projections to serotonergic neurons in the DR, which express both subtypes of orexin receptors (Mieda et al., 2011), while serotonin (5-HT) potently inhibits orexin neurons through activation of 5HT1A receptors (5HT1ARs). Serotonin 154-163 hypocretin Mus musculus 189-195 30348769-4 2018 With injection of adeno-associated virus into the orexin-Cre transgenic mouse brain, we expressed the DREADD receptor hM3Dq specifically in orexin neurons and applied the hM3Dq ligand clozapine to activate orexin neurons. Clozapine 184-193 hypocretin Mus musculus 50-56 30348769-4 2018 With injection of adeno-associated virus into the orexin-Cre transgenic mouse brain, we expressed the DREADD receptor hM3Dq specifically in orexin neurons and applied the hM3Dq ligand clozapine to activate orexin neurons. Clozapine 184-193 hypocretin Mus musculus 140-146 30348769-4 2018 With injection of adeno-associated virus into the orexin-Cre transgenic mouse brain, we expressed the DREADD receptor hM3Dq specifically in orexin neurons and applied the hM3Dq ligand clozapine to activate orexin neurons. Clozapine 184-193 hypocretin Mus musculus 140-146 30348769-7 2018 As an indication of reduced pain sensitivity, these orexin-DREADD mice took longer to respond to the 55 C thermal stimuli in the hot plate test and exhibited significantly less frequent licking of the formalin-injected paw in the formalin test. Formaldehyde 202-210 hypocretin Mus musculus 52-58 30348769-7 2018 As an indication of reduced pain sensitivity, these orexin-DREADD mice took longer to respond to the 55 C thermal stimuli in the hot plate test and exhibited significantly less frequent licking of the formalin-injected paw in the formalin test. Formaldehyde 231-239 hypocretin Mus musculus 52-58 30416119-8 2018 Additionally, YNT-185 promoted wakefulness in wild-type mice, suggesting that orexin receptor agonists may be useful for the treatment of excessive daytime sleepiness due to other conditions, such as sleepiness accompanying depression and sleepiness due to side effects of medicines or jet lag/shift work. YNT-185 14-21 hypocretin Mus musculus 78-84 30524223-6 2018 The results from animal experiments demonstrated that orexin-A attenuated the loss of dopaminergic neurons and the decrease of tyrosine hydroxylase (TH) expression in the substantia nigra, normalized the striatal dopaminergic fibers, and prevented the depletion of dopamine and its metabolites in the striatum. Dopamine 86-94 hypocretin Mus musculus 54-62 30524223-8 2018 Orexin-A improved MPTP-induced impairments in both motor activity and spatial memory. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 18-22 hypocretin Mus musculus 0-8 30524223-10 2018 Furthermore, the protective effects of orexin-A on MPTP parkinsonian mice could be blocked by orexinergic receptor 1 (OX1R) antagonist, SB334867. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 136-144 hypocretin Mus musculus 39-47 30524223-13 2018 The present study demonstrated that orexin-A exerted neuroprotective effects on MPTP parkinsonian mice, which may imply orexin-A as a potential therapeutic target for PD. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 80-84 hypocretin Mus musculus 36-44 30524223-13 2018 The present study demonstrated that orexin-A exerted neuroprotective effects on MPTP parkinsonian mice, which may imply orexin-A as a potential therapeutic target for PD. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 80-84 hypocretin Mus musculus 120-128 29557083-0 2018 SB-334867 (an Orexin-1 Receptor Antagonist) Effects on Morphine-Induced Sensitization in Mice-a View on Receptor Mechanisms. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 0-9 hypocretin Mus musculus 14-20 29557083-0 2018 SB-334867 (an Orexin-1 Receptor Antagonist) Effects on Morphine-Induced Sensitization in Mice-a View on Receptor Mechanisms. Morphine 55-63 hypocretin Mus musculus 14-20 29557083-1 2018 The present study focused upon the role of SB-334867, an orexin-1 receptor antagonist, in the acquisition of morphine-induced sensitization to locomotor activity in mice. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 43-52 hypocretin Mus musculus 57-63 29557083-1 2018 The present study focused upon the role of SB-334867, an orexin-1 receptor antagonist, in the acquisition of morphine-induced sensitization to locomotor activity in mice. Morphine 109-117 hypocretin Mus musculus 57-63 29557083-11 2018 Summing up, the orexin system may be an effective measure to inhibit morphine-induced behavioral sensitization. Morphine 69-77 hypocretin Mus musculus 16-22 30293961-0 2018 Relationship between the fluorescence intensity of rhodamine-labeled orexin A and the calcium responses in cortical neurons: An in vivo two-photon calcium imaging study. Rhodamines 51-60 hypocretin Mus musculus 69-77 29654707-0 2018 Melatonin promotes sleep in mice by inhibiting orexin neurons in the perifornical lateral hypothalamus. Melatonin 0-9 hypocretin Mus musculus 47-53 29654707-4 2018 Does melatonin promote sleep by inhibiting orexin neurons? Melatonin 5-14 hypocretin Mus musculus 43-49 29654707-12 2018 Local melatonin infusion at dark onset inhibited orexin neurons as evident by a significant reduction (66%, P = .0004) in the number of orexin neurons expressing c-Fos. Melatonin 6-15 hypocretin Mus musculus 49-55 29654707-12 2018 Local melatonin infusion at dark onset inhibited orexin neurons as evident by a significant reduction (66%, P = .0004) in the number of orexin neurons expressing c-Fos. Melatonin 6-15 hypocretin Mus musculus 136-142 29654707-14 2018 Based on these results, we suggest that melatonin may act via the MT1 receptors to inhibit orexin neurons and promote sleep. Melatonin 40-49 hypocretin Mus musculus 91-97 30293961-0 2018 Relationship between the fluorescence intensity of rhodamine-labeled orexin A and the calcium responses in cortical neurons: An in vivo two-photon calcium imaging study. Calcium 86-93 hypocretin Mus musculus 69-77 30293961-0 2018 Relationship between the fluorescence intensity of rhodamine-labeled orexin A and the calcium responses in cortical neurons: An in vivo two-photon calcium imaging study. Calcium 147-154 hypocretin Mus musculus 69-77 30293961-3 2018 Here, we synthesized a rhodamine-labeled orexin A compound, enabling us to quantify the amount of orexin binding to its receptors, OX1 and OX2, which principally couple to the Gq/11 protein. Rhodamines 23-32 hypocretin Mus musculus 41-49 30293961-3 2018 Here, we synthesized a rhodamine-labeled orexin A compound, enabling us to quantify the amount of orexin binding to its receptors, OX1 and OX2, which principally couple to the Gq/11 protein. Rhodamines 23-32 hypocretin Mus musculus 41-47 30293961-5 2018 Applying rhodamine-labeled orexin A (10 muM) to the cortical surface gradually and heterogeneously increased both the intensity of the rhodamine fluorescence and [Ca2+]i. Rhodamines 9-18 hypocretin Mus musculus 27-35 30293961-5 2018 Applying rhodamine-labeled orexin A (10 muM) to the cortical surface gradually and heterogeneously increased both the intensity of the rhodamine fluorescence and [Ca2+]i. Rhodamines 135-144 hypocretin Mus musculus 27-35 30293961-6 2018 Calcium responses started simultaneously with the increase in rhodamine-labeled orexin fluorescence and reached a plateau within several minutes. Calcium 0-7 hypocretin Mus musculus 80-86 30293961-6 2018 Calcium responses started simultaneously with the increase in rhodamine-labeled orexin fluorescence and reached a plateau within several minutes. Rhodamines 62-71 hypocretin Mus musculus 80-86 29950444-0 2018 Opiates increase the number of hypocretin-producing cells in human and mouse brain and reverse cataplexy in a mouse model of narcolepsy. Opiate Alkaloids 0-7 hypocretin Mus musculus 31-41 30225361-5 2018 To evaluate the relationship to the Hcrt system, we repeated the study in Orexin-tTA mice in the presence or absence of dietary doxycycline (DOX), which enabled us to manipulate the percentage of Hcrt neurons that expressed hM3Dq. hm3dq 224-229 hypocretin Mus musculus 196-200 30225361-6 2018 In DOX-fed mice, 18% of Hcrt neurons as well as many other LH neurons expressed hM3Dq; these mice showed a profound increase in wake after hM3Dq activation even in the presence of ALM. Doxycycline 3-6 hypocretin Mus musculus 24-28 29999239-4 2018 Importantly, we found that orexin-A accelerated osteoblast differentiation and matrix mineralization in MC3T3-E1 cells, as manifested by elevation of physiological markers of osteoblastic differentiation [alkaline phosphatase (ALP) and osteogenic genes] and Alizarin Red staining, respectively. alizarin 258-270 hypocretin Mus musculus 27-35 29999239-7 2018 Also, orexin-induced increase in gene expression (Runx-2, ALP, osteocalcin, and osterix) and matrix mineralization were prevented by the p38 MAPK specific inhibitor SB203580. SB 203580 165-173 hypocretin Mus musculus 6-12 29950444-4 2018 Studying five postmortem brains from heroin addicts, we report that the brain tissue had, on average, 54% more immunohistochemically detected neurons producing hypocretin than did control brains from neurologically normal subjects. Heroin 37-43 hypocretin Mus musculus 160-170 29950444-5 2018 Similar increases in hypocretin-producing cells could be induced in wild-type mice by long-term (but not short-term) administration of morphine. Morphine 135-143 hypocretin Mus musculus 21-31 29950444-8 2018 Morphine administration restored the population of detected hypocretin cells to normal numbers in transgenic mice in which these neurons had been partially depleted. Morphine 0-8 hypocretin Mus musculus 60-70 29950444-10 2018 These findings suggest that opiate agonists may have a role in the treatment of narcolepsy, a disorder caused by hypocretin neuron loss, and that increased numbers of hypocretin-producing cells may play a role in maintaining opiate addiction. Opiate Alkaloids 28-34 hypocretin Mus musculus 113-123 29315422-1 2018 Study Objectives: In previous work, dietary branched-chain amino acid (BCAA) supplementation, precursors to de novo glutamate and gamma-aminobutyric acid (GABA) synthesis, restored impaired sleep-wake regulation and orexin neuronal activity following traumatic brain injury (TBI) in mice. Amino Acids, Branched-Chain 44-69 hypocretin Mus musculus 216-222 29409952-10 2018 Moreover, the increased adenosine in KO LH contributed to lower spontaneous firing rates of putative wake-promoting orexin/hypocretin neurons. Adenosine 24-33 hypocretin Mus musculus 116-122 30155524-2 2018 To investigate co-expression of vesicular GABA transporter (VGAT, a marker of GABA neurons) and the vesicular glutamate transporter-2 (VGLUT2, a marker of glutamate neurons) in orexin and MCH neurons, we generated two transgenic mouse lines. gamma-Aminobutyric Acid 42-46 hypocretin Mus musculus 177-183 30155524-2 2018 To investigate co-expression of vesicular GABA transporter (VGAT, a marker of GABA neurons) and the vesicular glutamate transporter-2 (VGLUT2, a marker of glutamate neurons) in orexin and MCH neurons, we generated two transgenic mouse lines. Glutamic Acid 110-119 hypocretin Mus musculus 177-183 29454841-3 2018 We report here an extensive study on cocaine-related behaviors in hypocretin/orexin-deficient mice (KO) and their heterozygous (HET) and wildtype (WT) littermates. Cocaine 37-44 hypocretin Mus musculus 77-83 29454841-6 2018 Our observations show that whereas all mice exhibited quite similar responses to acute administration of cocaine, only Hcrt KO mice exhibited reduced cocaine-seeking behaviors following a period of abstinence or extinction, and reduced cocaine incubation craving. Cocaine 150-157 hypocretin Mus musculus 119-123 29454841-8 2018 We thus report that innate disruption of hypocretin/orexin signaling moderately alters cocaine reward but significantly reduces long-term affective dependence that may explain the lack of relapse for cocaine seeking seen in Hcrt KO mice. Cocaine 87-94 hypocretin Mus musculus 52-58 29315422-1 2018 Study Objectives: In previous work, dietary branched-chain amino acid (BCAA) supplementation, precursors to de novo glutamate and gamma-aminobutyric acid (GABA) synthesis, restored impaired sleep-wake regulation and orexin neuronal activity following traumatic brain injury (TBI) in mice. Amino Acids, Branched-Chain 71-75 hypocretin Mus musculus 216-222 29315422-2 2018 TBI was speculated to reduce orexin neuronal activity through decreased regional excitatory (glutamate) and/or increased inhibitory (GABA) input. Glutamic Acid 93-102 hypocretin Mus musculus 29-35 29315422-2 2018 TBI was speculated to reduce orexin neuronal activity through decreased regional excitatory (glutamate) and/or increased inhibitory (GABA) input. gamma-Aminobutyric Acid 133-137 hypocretin Mus musculus 29-35 29315422-3 2018 Therefore, we hypothesized that TBI would decrease synaptic glutamate and/or increase synaptic GABA in nerve terminals contacting orexin neurons, and BCAA supplementation would restore TBI-induced changes in synaptic glutamate and/or GABA. gamma-Aminobutyric Acid 95-99 hypocretin Mus musculus 130-136 29315422-8 2018 However, BCAA supplementation only restored relative glutamate density within presynaptic terminals contacting orexin-positive hypothalamic neurons. Amino Acids, Branched-Chain 9-13 hypocretin Mus musculus 111-117 29315422-10 2018 Conclusions: These results suggest TBI compromises orexin neuron function via decreased glutamate density and highlight BCAA supplementation as a potential therapy to restore glutamate density to orexin neurons. Amino Acids, Branched-Chain 120-124 hypocretin Mus musculus 196-202 29315422-10 2018 Conclusions: These results suggest TBI compromises orexin neuron function via decreased glutamate density and highlight BCAA supplementation as a potential therapy to restore glutamate density to orexin neurons. Glutamic Acid 175-184 hypocretin Mus musculus 196-202 29311142-7 2018 Photostimulation of LH/PF GABAergic neurons inhibited the firing of orexin neurons through the release of GABA, evoking GABAA-mediated IPSCs in orexin neurons. gamma-Aminobutyric Acid 26-30 hypocretin Mus musculus 68-74 29311142-7 2018 Photostimulation of LH/PF GABAergic neurons inhibited the firing of orexin neurons through the release of GABA, evoking GABAA-mediated IPSCs in orexin neurons. gamma-Aminobutyric Acid 26-30 hypocretin Mus musculus 144-150 29311142-12 2018 We propose that local release of dynorphin may, via collaterals, provides a positive feedback to orexin neurons and that, during wakefulness, orexin neurons may be disinhibited by acetylcholine and by their own release of dynorphin.SIGNIFICANCE STATEMENT The lateral hypothalamus contains important wake-promoting cell populations, including orexin-producing neurons. Acetylcholine 180-193 hypocretin Mus musculus 142-148 29311142-12 2018 We propose that local release of dynorphin may, via collaterals, provides a positive feedback to orexin neurons and that, during wakefulness, orexin neurons may be disinhibited by acetylcholine and by their own release of dynorphin.SIGNIFICANCE STATEMENT The lateral hypothalamus contains important wake-promoting cell populations, including orexin-producing neurons. Acetylcholine 180-193 hypocretin Mus musculus 142-148 28549585-0 2017 Cystamine-mediated inhibition of protein disulfide isomerase triggers aggregation of misfolded orexin-A in the Golgi apparatus and prevents extracellular secretion of orexin-A. Cystamine 0-9 hypocretin Mus musculus 95-103 29426934-0 2018 Involvement of orexin neurons in fasting- and central adenosine-induced hypothermia. Adenosine 54-63 hypocretin Mus musculus 15-21 29426934-1 2018 We examined whether orexin neurons might play a protective role against fasting- and adenosine-induced hypothermia. Adenosine 85-94 hypocretin Mus musculus 20-26 29426934-4 2018 Orexin neurons were active just before onset of hypothermia and during the recovery period as revealed by calcium imaging in vivo using G-CaMP. Calcium 106-113 hypocretin Mus musculus 0-6 29426934-4 2018 Orexin neurons were active just before onset of hypothermia and during the recovery period as revealed by calcium imaging in vivo using G-CaMP. g-camp 136-142 hypocretin Mus musculus 0-6 29426934-9 2018 We propose that orexin neurons play dual roles (enhancer in the induction phase and compensator during the recovery phase) in adenosine-induced hypothermia and a protective/compensatory role in fasting-induced hypothermia. Adenosine 126-135 hypocretin Mus musculus 16-22 28966976-7 2017 Virtually all MCH and approximately half of the Hcrt/Ox neurons sampled express both the machinery for glutamate release and GABA synthesis in the absence of a vesicular GABA release pathway. Glutamic Acid 103-112 hypocretin Mus musculus 48-52 28966976-7 2017 Virtually all MCH and approximately half of the Hcrt/Ox neurons sampled express both the machinery for glutamate release and GABA synthesis in the absence of a vesicular GABA release pathway. gamma-Aminobutyric Acid 125-129 hypocretin Mus musculus 48-52 28867552-5 2017 This astrocyte-specific genetic manipulation silenced the wake-promoting orexin neurons located in the lateral hypothalamic area (LHA) by impairing glucose and lactate trafficking through astrocytic networks. Glucose 148-155 hypocretin Mus musculus 73-79 28867552-5 2017 This astrocyte-specific genetic manipulation silenced the wake-promoting orexin neurons located in the lateral hypothalamic area (LHA) by impairing glucose and lactate trafficking through astrocytic networks. Lactic Acid 160-167 hypocretin Mus musculus 73-79 28877723-9 2017 Besides, this CPP training process-induced hippocampal re-potentiation was prevented when mice were pretreated with TCS1102, a dual orexin receptor antagonist. TCS 1102 116-123 hypocretin Mus musculus 132-138 29038792-0 2017 Neuroanatomical Relationships between Orexin/Hypocretin-Containing Neurons/Nerve Fibers and Nicotine-Induced c-Fos-Activated Cells of the Reward-Addiction Neurocircuitry. Nicotine 92-100 hypocretin Mus musculus 38-44 29038792-0 2017 Neuroanatomical Relationships between Orexin/Hypocretin-Containing Neurons/Nerve Fibers and Nicotine-Induced c-Fos-Activated Cells of the Reward-Addiction Neurocircuitry. Nicotine 92-100 hypocretin Mus musculus 45-55 29038792-1 2017 Orexin/hypocretin-containing neurons in lateral hypothalamus (LH) are implicated in the neurobiology of nicotine addiction. Nicotine 104-112 hypocretin Mus musculus 0-6 29038792-1 2017 Orexin/hypocretin-containing neurons in lateral hypothalamus (LH) are implicated in the neurobiology of nicotine addiction. Nicotine 104-112 hypocretin Mus musculus 7-17 28549585-3 2017 Orexin-A has two closely located intramolecular disulfide bonds and is prone to misfolding due to the formation of incorrect disulfide bonds. Disulfides 48-57 hypocretin Mus musculus 0-8 28549585-3 2017 Orexin-A has two closely located intramolecular disulfide bonds and is prone to misfolding due to the formation of incorrect disulfide bonds. Disulfides 125-134 hypocretin Mus musculus 0-8 28549585-5 2017 PDI can modify misfolded orexin-A to its native form by rearrangement of two disulfide bonds. Disulfides 77-86 hypocretin Mus musculus 25-33 28549585-11 2017 Moreover, cystamine triggered aggregation of orexin-A, but not orexin-B in the Golgi apparatus of hypothalamic slice cultures and in vivo mouse brains. Cystamine 10-19 hypocretin Mus musculus 45-53 28549585-13 2017 Finally, we demonstrated that cystamine significantly decreased extracellular secretion of orexin-A in AD293 cells overexpressing prepro-orexin. Cystamine 30-39 hypocretin Mus musculus 91-99 30504686-0 2018 Endogenous Nitric Oxide Inhibits, Whereas Awakening Stimuli Increase, the Activity of a Subset of Orexin Neurons. Nitric Oxide 11-23 hypocretin Mus musculus 98-104 30504686-2 2018 Here we examined whether the activity of orexin neurons was regulated by endogenous nitric oxide (NO) in male C57BL/6 mice. Nitric Oxide 84-96 hypocretin Mus musculus 41-47 27480648-0 2017 Hypocretin/orexin knock-out mice display disrupted behavioral and dopamine responses to cocaine. Dopamine 66-74 hypocretin Mus musculus 0-17 27480648-0 2017 Hypocretin/orexin knock-out mice display disrupted behavioral and dopamine responses to cocaine. Cocaine 88-95 hypocretin Mus musculus 0-17 27480648-1 2017 The hypocretin/orexin (HCRT) system is implicated in reward and reinforcement processes through actions on the mesolimbic dopamine (DA) system. Dopamine 122-130 hypocretin Mus musculus 4-21 27480648-1 2017 The hypocretin/orexin (HCRT) system is implicated in reward and reinforcement processes through actions on the mesolimbic dopamine (DA) system. Dopamine 122-130 hypocretin Mus musculus 23-27 27480648-1 2017 The hypocretin/orexin (HCRT) system is implicated in reward and reinforcement processes through actions on the mesolimbic dopamine (DA) system. Dopamine 132-134 hypocretin Mus musculus 4-21 27480648-1 2017 The hypocretin/orexin (HCRT) system is implicated in reward and reinforcement processes through actions on the mesolimbic dopamine (DA) system. Dopamine 132-134 hypocretin Mus musculus 23-27 27480648-2 2017 Here we provide evidence for the relationship between HCRT and DA in vivo in anesthetized and freely moving mice. Dopamine 63-65 hypocretin Mus musculus 54-58 27480648-4 2017 Using a combination of microdialysis and in vivo fast scan cyclic voltammetry in anesthetized and freely moving mice, we investigated the underlying role of HCRT in the regulation of DA release and uptake. Dopamine 183-185 hypocretin Mus musculus 157-161 27480648-7 2017 Further, diminished DA signaling in HCRT KO mice persists following administration of cocaine. Cocaine 86-93 hypocretin Mus musculus 36-40 27480648-8 2017 These findings indicate that HCRT is essential for the expression of behaviors associated with the rewarding effects of cocaine, and suggest that HCRT regulation of reward and reinforcement may be related to disruptions to DA neurotransmission. Cocaine 120-127 hypocretin Mus musculus 29-33 28778226-3 2017 Although hypocretin receptor antagonists have been developed as sleep-inducing drugs, a high dose of suvorexant, a hypocretin receptor antagonist, inhibits gene expression of prepro-hypocretin to induce narcoleptic attack in wild-type mice. suvorexant 101-111 hypocretin Mus musculus 9-19 28778226-3 2017 Although hypocretin receptor antagonists have been developed as sleep-inducing drugs, a high dose of suvorexant, a hypocretin receptor antagonist, inhibits gene expression of prepro-hypocretin to induce narcoleptic attack in wild-type mice. suvorexant 101-111 hypocretin Mus musculus 115-125 28778226-3 2017 Although hypocretin receptor antagonists have been developed as sleep-inducing drugs, a high dose of suvorexant, a hypocretin receptor antagonist, inhibits gene expression of prepro-hypocretin to induce narcoleptic attack in wild-type mice. suvorexant 101-111 hypocretin Mus musculus 115-125 28453642-5 2017 Results: Treatment with the orexin-1 receptor antagonist SB334867 increased the activity of basolateral amygdala neurons projecting to infralimbic medial prefrontal cortex during fear extinction. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 57-65 hypocretin Mus musculus 28-34 28453642-7 2017 Conclusions: These data identify neuronal circuits and cell populations of the amygdala associated with the facilitation of fear extinction consolidation induced by the orexin-1 receptor antagonist SB334867. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 198-206 hypocretin Mus musculus 169-175 28549585-14 2017 These findings suggest that cystamine-induced PDI inhibition induces selective depletion, aggregation in the Golgi apparatus and impaired secretion of orexin-A. Cystamine 28-37 hypocretin Mus musculus 151-159 28549585-0 2017 Cystamine-mediated inhibition of protein disulfide isomerase triggers aggregation of misfolded orexin-A in the Golgi apparatus and prevents extracellular secretion of orexin-A. Cystamine 0-9 hypocretin Mus musculus 167-175 28353077-5 2017 RECENT FINDINGS: In normal rats and mice, central administration of orexin increases food intake, blood pressure, and sympathetic nerve activity and these effects are blocked by selective orexin receptor antagonist SB-334867 or almorexant. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 215-224 hypocretin Mus musculus 68-74 28363838-13 2017 Then, among animals on HFD, orexin neurons were activated following injections of clozapine n-oxide (CNO). clozapine N-oxide 82-99 hypocretin Mus musculus 28-34 28507129-5 2017 Intraperitoneal, and intracerebroventricular, administration of YNT-185 suppressed cataplexy-like episodes in orexin knockout and orexin neuron-ablated mice, but not in orexin receptor-deficient mice. YNT-185 64-71 hypocretin Mus musculus 110-116 28507129-5 2017 Intraperitoneal, and intracerebroventricular, administration of YNT-185 suppressed cataplexy-like episodes in orexin knockout and orexin neuron-ablated mice, but not in orexin receptor-deficient mice. YNT-185 64-71 hypocretin Mus musculus 130-136 28507129-5 2017 Intraperitoneal, and intracerebroventricular, administration of YNT-185 suppressed cataplexy-like episodes in orexin knockout and orexin neuron-ablated mice, but not in orexin receptor-deficient mice. YNT-185 64-71 hypocretin Mus musculus 130-136 28580289-2 2017 The altered OX level is linked to a dramatic increase of the inhibitory innervation of OX producing neurons (OX neurons) and the over expression of the endocannabinoid 2-arachidonoylglycerol (2-AG) by OX neurons of ob/ob mice. Endocannabinoids 152-167 hypocretin Mus musculus 12-14 28580289-2 2017 The altered OX level is linked to a dramatic increase of the inhibitory innervation of OX producing neurons (OX neurons) and the over expression of the endocannabinoid 2-arachidonoylglycerol (2-AG) by OX neurons of ob/ob mice. glyceryl 2-arachidonate 168-190 hypocretin Mus musculus 12-14 28580289-2 2017 The altered OX level is linked to a dramatic increase of the inhibitory innervation of OX producing neurons (OX neurons) and the over expression of the endocannabinoid 2-arachidonoylglycerol (2-AG) by OX neurons of ob/ob mice. glyceryl 2-arachidonate 192-196 hypocretin Mus musculus 12-14 28580289-10 2017 Finally, the physiologic release of 2-AG induces a prevalent depression of inhibition (disinhibition) of OX neurons in ob/ob animals but not in wt. glyceryl 2-arachidonate 36-40 hypocretin Mus musculus 105-107 28580289-11 2017 CONCLUSIONS: In ob/ob mice, chronic absence of leptin induces a 2-AG mediated functional disinhibition of OX neurons. glyceryl 2-arachidonate 64-68 hypocretin Mus musculus 106-108 28533747-2 2017 Stress or orexin administration stimulates hyperarousal, adrenocorticotropic hormone (ACTH) and corticosterone release, and selective OX1R blockade can attenuate several stress-induced behavioral and cardiovascular responses but not the hypothalamic-pituitary-adrenal (HPA) axis activation. Corticosterone 96-110 hypocretin Mus musculus 10-16 28396432-0 2017 Serotonin neurons in the dorsal raphe mediate the anticataplectic action of orexin neurons by reducing amygdala activity. Serotonin 0-9 hypocretin Mus musculus 76-82 28396432-2 2017 In a mouse model for narcolepsy, we previously demonstrated that serotonin neurons of the dorsal raphe nucleus (DRN) mediate the suppression of cataplexy-like episodes (CLEs) by orexin neurons. Serotonin 65-74 hypocretin Mus musculus 178-184 28396432-5 2017 Moreover, the optogenetic inhibition of serotonin nerve terminals in the amygdala blocked the anticataplectic effects of orexin signaling in DRN serotonin neurons. Serotonin 40-49 hypocretin Mus musculus 121-127 28396432-5 2017 Moreover, the optogenetic inhibition of serotonin nerve terminals in the amygdala blocked the anticataplectic effects of orexin signaling in DRN serotonin neurons. Serotonin 145-154 hypocretin Mus musculus 121-127 28396432-6 2017 Taken together, the results suggest that DRN serotonin neurons, as a downstream target of orexin neurons, inhibit cataplexy by reducing the activity of amygdala as a center for emotional processing. Serotonin 45-54 hypocretin Mus musculus 90-96 28223018-0 2017 Cerebral ischemia-induced elevation of hepatic inflammatory factors accompanied by glucose intolerance suppresses hypothalamic orexin-A-mediated vagus nerve activation. Glucose 83-90 hypocretin Mus musculus 127-135 28209737-8 2017 To manipulate GABA cells specifically, we developed a new mouse line that enables genetic targeting of GABA cells in orexin-/- mice. gamma-Aminobutyric Acid 103-107 hypocretin Mus musculus 117-123 28353077-5 2017 RECENT FINDINGS: In normal rats and mice, central administration of orexin increases food intake, blood pressure, and sympathetic nerve activity and these effects are blocked by selective orexin receptor antagonist SB-334867 or almorexant. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 215-224 hypocretin Mus musculus 188-194 28041630-9 2017 Intracerebroventricularly administered orexin-A dose-dependently attenuated oxaliplatin-induced mechanical allodynia and increased tail flick latencies. Oxaliplatin 76-87 hypocretin Mus musculus 39-47 28178514-0 2017 Projection-Target-Defined Effects of Orexin and Dynorphin on VTA Dopamine Neurons. Dopamine 65-73 hypocretin Mus musculus 37-43 28178514-2 2017 We tested the actions of co-expressed lateral hypothalamic peptides, orexin A (oxA) and dynorphin (dyn), on projection-target-defined dopamine neurons in mice. Dopamine 134-142 hypocretin Mus musculus 69-77 28178514-5 2017 OxA selectively increased firing in lAcbSh- and mAcbSh-projecting dopamine neurons. lacbsh 36-42 hypocretin Mus musculus 0-3 28178514-5 2017 OxA selectively increased firing in lAcbSh- and mAcbSh-projecting dopamine neurons. Dopamine 66-74 hypocretin Mus musculus 0-3 28097729-0 2017 The Role of Orexin Signaling in the Ventral Tegmental Area and Central Amygdala in Modulating Binge-Like Ethanol Drinking Behavior. Ethanol 105-112 hypocretin Mus musculus 12-18 28097729-1 2017 BACKGROUND: Recent reports have demonstrated that binge-like ethanol (EtOH) drinking leads to an increase in hypothalamic orexin (OX) signaling and that suppressing this signaling via systemic administration of an orexin receptor (OXR) antagonist blocks this behavior; however, the specific OX pathways that modulate this behavior remain unknown. Ethanol 61-68 hypocretin Mus musculus 122-128 28097729-1 2017 BACKGROUND: Recent reports have demonstrated that binge-like ethanol (EtOH) drinking leads to an increase in hypothalamic orexin (OX) signaling and that suppressing this signaling via systemic administration of an orexin receptor (OXR) antagonist blocks this behavior; however, the specific OX pathways that modulate this behavior remain unknown. Ethanol 61-68 hypocretin Mus musculus 130-132 28097729-1 2017 BACKGROUND: Recent reports have demonstrated that binge-like ethanol (EtOH) drinking leads to an increase in hypothalamic orexin (OX) signaling and that suppressing this signaling via systemic administration of an orexin receptor (OXR) antagonist blocks this behavior; however, the specific OX pathways that modulate this behavior remain unknown. Ethanol 61-68 hypocretin Mus musculus 214-220 28097729-1 2017 BACKGROUND: Recent reports have demonstrated that binge-like ethanol (EtOH) drinking leads to an increase in hypothalamic orexin (OX) signaling and that suppressing this signaling via systemic administration of an orexin receptor (OXR) antagonist blocks this behavior; however, the specific OX pathways that modulate this behavior remain unknown. Ethanol 61-68 hypocretin Mus musculus 231-233 28097729-1 2017 BACKGROUND: Recent reports have demonstrated that binge-like ethanol (EtOH) drinking leads to an increase in hypothalamic orexin (OX) signaling and that suppressing this signaling via systemic administration of an orexin receptor (OXR) antagonist blocks this behavior; however, the specific OX pathways that modulate this behavior remain unknown. Ethanol 70-74 hypocretin Mus musculus 122-128 28097729-1 2017 BACKGROUND: Recent reports have demonstrated that binge-like ethanol (EtOH) drinking leads to an increase in hypothalamic orexin (OX) signaling and that suppressing this signaling via systemic administration of an orexin receptor (OXR) antagonist blocks this behavior; however, the specific OX pathways that modulate this behavior remain unknown. Ethanol 70-74 hypocretin Mus musculus 130-132 28097729-1 2017 BACKGROUND: Recent reports have demonstrated that binge-like ethanol (EtOH) drinking leads to an increase in hypothalamic orexin (OX) signaling and that suppressing this signaling via systemic administration of an orexin receptor (OXR) antagonist blocks this behavior; however, the specific OX pathways that modulate this behavior remain unknown. Ethanol 70-74 hypocretin Mus musculus 214-220 28097729-1 2017 BACKGROUND: Recent reports have demonstrated that binge-like ethanol (EtOH) drinking leads to an increase in hypothalamic orexin (OX) signaling and that suppressing this signaling via systemic administration of an orexin receptor (OXR) antagonist blocks this behavior; however, the specific OX pathways that modulate this behavior remain unknown. Ethanol 70-74 hypocretin Mus musculus 231-233 28097729-2 2017 The goal of this study was to further elucidate the role of the OX system in binge-like EtOH drinking using behavioral, molecular, and pharmacological techniques. Ethanol 88-92 hypocretin Mus musculus 64-66 28097729-4 2017 Experiment 1 examined changes in the OX precursor, prepro-orexin, within the hypothalamus following multiple cycle EtOH or sucrose DID using polymerase chain reaction (PCR) analysis. Ethanol 115-119 hypocretin Mus musculus 37-39 28097729-4 2017 Experiment 1 examined changes in the OX precursor, prepro-orexin, within the hypothalamus following multiple cycle EtOH or sucrose DID using polymerase chain reaction (PCR) analysis. Ethanol 115-119 hypocretin Mus musculus 51-64 28097729-4 2017 Experiment 1 examined changes in the OX precursor, prepro-orexin, within the hypothalamus following multiple cycle EtOH or sucrose DID using polymerase chain reaction (PCR) analysis. Sucrose 123-130 hypocretin Mus musculus 37-39 28097729-4 2017 Experiment 1 examined changes in the OX precursor, prepro-orexin, within the hypothalamus following multiple cycle EtOH or sucrose DID using polymerase chain reaction (PCR) analysis. Sucrose 123-130 hypocretin Mus musculus 51-64 28097729-8 2017 Interestingly, inhibition of OX1Rs did not affect binge-like sucrose intake, which suggests that these OX circuits are specific for EtOH consumption. Ethanol 132-136 hypocretin Mus musculus 29-31 28097729-9 2017 CONCLUSIONS: As a whole, these data suggest that the VTA and CeA are important regions in which OX regulates binge-like EtOH drinking behavior. Ethanol 120-124 hypocretin Mus musculus 96-98 28041630-10 2017 Oxaliplatin-induced mechanical allodynia was completely reversed by orexin-A at a low dose that did not increase tail flick latency. Oxaliplatin 0-11 hypocretin Mus musculus 68-76 28041630-12 2017 The analgesic effect of orexin-A on oxaliplatin-induced mechanical allodynia was completely antagonized by prior intraperitoneal injection of SB-408124 (orexin type-1 receptor antagonist), but not by prior intraperitoneal injection of TCS-OX2-29 (orexin type-2 receptor antagonist). Oxaliplatin 36-47 hypocretin Mus musculus 24-32 28041630-12 2017 The analgesic effect of orexin-A on oxaliplatin-induced mechanical allodynia was completely antagonized by prior intraperitoneal injection of SB-408124 (orexin type-1 receptor antagonist), but not by prior intraperitoneal injection of TCS-OX2-29 (orexin type-2 receptor antagonist). SB 408124 142-151 hypocretin Mus musculus 24-32 28041630-12 2017 The analgesic effect of orexin-A on oxaliplatin-induced mechanical allodynia was completely antagonized by prior intraperitoneal injection of SB-408124 (orexin type-1 receptor antagonist), but not by prior intraperitoneal injection of TCS-OX2-29 (orexin type-2 receptor antagonist). 1-(3,4-dihydro-6,7-dimethoxy-2(1H)-isoquinolinyl)-3,3-dimethyl-2-((4-pyridinylmethyl)amino)-1-butanone 235-245 hypocretin Mus musculus 24-32 27174745-6 2017 In testicular sections on 0day postpartum (dpp), gonocytes, Sertoli cells and foetal Leydig cells showed OXA and OX1R-immunopositive signals. dipalmitoylphosphatidylserine 43-46 hypocretin Mus musculus 105-108 26562300-6 2016 For this, binding of OXA to OX1R was blocked using OX1R specific antagonist, SB-334867. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 77-86 hypocretin Mus musculus 21-24 26562300-10 2016 The decrease in expressions of OXA, OX1R and GLUT 3 in the test is in response to both doses of the antagonist points to their down-regulation causing inefficient uptake of glucose by the testicular cells, thereby affecting gonadal development. Glucose 173-180 hypocretin Mus musculus 31-34 27751532-0 2016 Blockade of dorsal hippocampal orexin-1 receptors impaired morphine-induced state-dependent learning. Morphine 59-67 hypocretin Mus musculus 31-37 27751532-6 2016 Pre-test administration of orexin-1 (0.5, 5 and 50pmol, intra-CA1) had not a significant effect on the retention latency compared to the saline-treated animals, but it restored the memory impairment induced by pre-training morphine (5mg/kg, i.p.). Morphine 223-231 hypocretin Mus musculus 27-33 27751532-10 2016 Conversely, pre-test injection of SB-334867 (10, 20 and 40nmol, intra-CA1) inhibited the orexin-1-induced potentiation of morphine state-dependent learning on the test day. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 34-43 hypocretin Mus musculus 89-95 27751532-10 2016 Conversely, pre-test injection of SB-334867 (10, 20 and 40nmol, intra-CA1) inhibited the orexin-1-induced potentiation of morphine state-dependent learning on the test day. Morphine 122-130 hypocretin Mus musculus 89-95 27751532-11 2016 It is concluded that dorsal hippocampal orexin-1 receptors may be involved, at least in part, in morphine state-dependent learning in mice. Morphine 97-105 hypocretin Mus musculus 40-46 27631554-2 2016 To investigate this possibility, we here examined the effects of suvorexant, an antiinsomnia drug targeting the orexin system, on sleep and glucose metabolism in type 2 diabetic mice. suvorexant 65-75 hypocretin Mus musculus 112-118 27845440-7 2016 Furthermore, immunohistochemical analysis revealed that linalool activated hypothalamic orexin neurons, one of the key mediators for pain processing. linalool 56-64 hypocretin Mus musculus 88-94 27845440-8 2016 Formalin tests in orexin neuron-ablated and orexin peptide-deficient mice showed orexinergic transmission was essential for linalool odour-induced analgesia. Formaldehyde 0-8 hypocretin Mus musculus 18-24 27230703-2 2016 The activity of histamine neurons is increased by orexin neurons. Histamine 16-25 hypocretin Mus musculus 50-56 27683906-3 2016 This orexin-enhanced AHP (oeAHP) was mediated by both OX1 and OX2 receptors, required Ca(2+) influx, reversed near EK, and decayed with two components, the faster of which resulted from enhanced SK channel activation, whereas the slower component decayed like a slow AHP (sAHP), but was not blocked by UCL2077, an antagonist of sAHPs in some neurons. sahps 328-333 hypocretin Mus musculus 5-11 27449757-0 2016 Orexin A attenuates palmitic acid-induced hypothalamic cell death. Palmitic Acid 20-33 hypocretin Mus musculus 0-8 27235100-10 2016 These results suggest that hcrt/orx, noradrenaline, acetylcholine, and neurotensin could contribute to high-frequency cortical activity through an action on L6b GABAergic FS cells. orx 32-35 hypocretin Mus musculus 27-31 27436148-5 2016 FRET and co-immunoprecipitation experiments showed the formation of OX-1R/CB1R heteromers after incubation with OX-A (0.2muM), or OX-A (0.1muM)+ACEA (0.1muM), but not after ACEA (0.2muM), in a manner antagonized by SB-334867 or AM251. arachidonyl-2-chloroethylamide 144-148 hypocretin Mus musculus 112-116 27436148-5 2016 FRET and co-immunoprecipitation experiments showed the formation of OX-1R/CB1R heteromers after incubation with OX-A (0.2muM), or OX-A (0.1muM)+ACEA (0.1muM), but not after ACEA (0.2muM), in a manner antagonized by SB-334867 or AM251. arachidonyl-2-chloroethylamide 144-148 hypocretin Mus musculus 130-134 27436148-5 2016 FRET and co-immunoprecipitation experiments showed the formation of OX-1R/CB1R heteromers after incubation with OX-A (0.2muM), or OX-A (0.1muM)+ACEA (0.1muM), but not after ACEA (0.2muM), in a manner antagonized by SB-334867 or AM251. arachidonyl-2-chloroethylamide 173-177 hypocretin Mus musculus 112-116 27436148-5 2016 FRET and co-immunoprecipitation experiments showed the formation of OX-1R/CB1R heteromers after incubation with OX-A (0.2muM), or OX-A (0.1muM)+ACEA (0.1muM), but not after ACEA (0.2muM), in a manner antagonized by SB-334867 or AM251. arachidonyl-2-chloroethylamide 173-177 hypocretin Mus musculus 130-134 27436148-5 2016 FRET and co-immunoprecipitation experiments showed the formation of OX-1R/CB1R heteromers after incubation with OX-A (0.2muM), or OX-A (0.1muM)+ACEA (0.1muM), but not after ACEA (0.2muM), in a manner antagonized by SB-334867 or AM251. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 215-224 hypocretin Mus musculus 112-116 27436148-5 2016 FRET and co-immunoprecipitation experiments showed the formation of OX-1R/CB1R heteromers after incubation with OX-A (0.2muM), or OX-A (0.1muM)+ACEA (0.1muM), but not after ACEA (0.2muM), in a manner antagonized by SB-334867 or AM251. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 215-224 hypocretin Mus musculus 130-134 27436148-6 2016 OX-A (0.2muM) or OX-A (0.1muM)+ACEA (0.1muM) also led to 2-AG biosynthesis. glyceryl 2-arachidonate 57-61 hypocretin Mus musculus 0-4 27436148-6 2016 OX-A (0.2muM) or OX-A (0.1muM)+ACEA (0.1muM) also led to 2-AG biosynthesis. glyceryl 2-arachidonate 57-61 hypocretin Mus musculus 17-21 27436148-7 2016 Finally, a stronger activation of ERK1/2(Thr202/185) phosphorylation in comparison to basal or each agonist alone (0.1-0.2muM), was induced by incubation with OX-A (0.1muM)+ACEA (0.1muM), again in a manner prevented by OX-1R or CB1R antagonism. arachidonyl-2-chloroethylamide 173-177 hypocretin Mus musculus 159-163 27625592-2 2016 Orexin-1-type receptors (OX1Rs) promote reward intake under conditions of strong drives for reward, including excessive alcohol intake. Alcohols 120-127 hypocretin Mus musculus 0-6 27625592-6 2016 In contrast, intra-mNAsh infusion of the orexin-2 receptor TCS-OX2-29 had no impact on alcohol drinking. Technetium 59-62 hypocretin Mus musculus 41-47 27625592-10 2016 Finally, orexin ex vivo enhanced firing in mNAsh neurons from alcohol-drinking mice, with no effect on evoked EPSCs or input resistance; a similar orexin increase in firing without a change in input resistance was observed in alcohol-naive mice. Alcohols 62-69 hypocretin Mus musculus 9-15 26965217-4 2016 Orexin A ( 30 nM) attenuated LTP induced by theta burst stimulation (TBS) in a manner antagonized by an OX1R (SB-334867), but not OX2R (EMPA), antagonist. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 110-119 hypocretin Mus musculus 0-8 27068481-0 2016 Narcolepsy-like sleep disturbance in orexin knockout mice are normalized by the 5-HT1A receptor agonist 8-OH-DPAT. 8-Hydroxy-2-(di-n-propylamino)tetralin 104-113 hypocretin Mus musculus 37-43 26907809-9 2016 [Ala(11), D-Leu(15)]-orexin B (i.pag. Alanine 1-4 hypocretin Mus musculus 21-29 26907809-9 2016 [Ala(11), D-Leu(15)]-orexin B (i.pag. D-LEUCINE 10-15 hypocretin Mus musculus 21-29 26907809-9 2016 [Ala(11), D-Leu(15)]-orexin B (i.pag. phenylacetylglycine 33-36 hypocretin Mus musculus 21-29 26907809-15 2016 These results suggest that during stress, hypothalamic orexin neurons are activated, releasing orexins into the vlPAG to induce analgesia, possibly via the OX1 receptor-initiated, endocannabinoid-mediated disinhibition mechanism previously reported. Endocannabinoids 180-195 hypocretin Mus musculus 55-61 27118715-5 2016 Orexin-deficient mice were treated with different doses of the norepinephrine reuptake inhibitor reboxetine, the serotonin reuptake inhibitor escitalopram, the alpha1 receptor agonist cirazoline or the alpha1 receptor antagonist prazosin. Reboxetine 97-107 hypocretin Mus musculus 0-6 27118715-8 2016 Reboxetine (doses >=0.55mg/kg) as well as escitalopram (doses >=3.0mg/kg) dose-dependently reduced the number of narcoleptic episodes in orexin-deficient mice. Reboxetine 0-10 hypocretin Mus musculus 143-149 27118715-8 2016 Reboxetine (doses >=0.55mg/kg) as well as escitalopram (doses >=3.0mg/kg) dose-dependently reduced the number of narcoleptic episodes in orexin-deficient mice. Citalopram 45-57 hypocretin Mus musculus 143-149 27294196-6 2016 Although, GABABRs were observed in all Orx and MCH neurons (100%), the luminance of these receptors was differentially altered following SD. gababrs 10-17 hypocretin Mus musculus 39-42 27294196-7 2016 The intensity of GABABRs in the Orx neurons was significantly greater after SD than after SC and SR, whereas that in the MCH neurons was significantly less. gababrs 17-24 hypocretin Mus musculus 32-35 27091533-9 2016 Co-administration of modafinil/flecainide resulted in a marked decrease in the number and duration of direct transitions to rapid eye movement sleep, which are characteristic of narcoleptic episodes in orexin knockout mice. Flecainide 31-41 hypocretin Mus musculus 202-208 27068481-3 2016 OBJECTIVES: The present study was designed to investigate both the nature of the neuronal changes in orexin KO mice and the therapeutic effects of monoamine-related compounds on the sleep disorder in orexin KO mice. monoamine 147-156 hypocretin Mus musculus 200-206 27068481-6 2016 In addition, the sleep disorder in orexin KO mice, as analyzed by a polysomnography during the dark period, was completely normalized by 8-OH-DPAT. 8-Hydroxy-2-(di-n-propylamino)tetralin 137-146 hypocretin Mus musculus 35-41 27276194-2 2016 Pharmacological treatment with the potent, selective and structurally distinct dual orexin receptor antagonists (ORAs) DORA-12 and DORA-2 significantly reduced pain responses during both phases I and II of the mouse formalin pain model and significantly reversed hyperalgesia in the rat complete Freund"s adjuvant pain model, respectively. Formaldehyde 216-224 hypocretin Mus musculus 84-90 27071101-0 2016 Orexin-A represses satiety-inducing POMC neurons and contributes to obesity via stimulation of endocannabinoid signaling. Endocannabinoids 95-110 hypocretin Mus musculus 0-8 27071101-3 2016 OX-A is crucial for the control of wakefulness and energy homeostasis and promotes, in OX-1R-expressing cells, the biosynthesis of the endogenous counterpart of marijuana"s psychotropic and appetite-inducing component Delta(9)-tetrahydrocannabinol, i.e., the endocannabinoid 2-arachidonoylglycerol (2-AG), which acts at CB1R. Dronabinol 218-247 hypocretin Mus musculus 0-4 27071101-3 2016 OX-A is crucial for the control of wakefulness and energy homeostasis and promotes, in OX-1R-expressing cells, the biosynthesis of the endogenous counterpart of marijuana"s psychotropic and appetite-inducing component Delta(9)-tetrahydrocannabinol, i.e., the endocannabinoid 2-arachidonoylglycerol (2-AG), which acts at CB1R. glyceryl 2-arachidonate 275-297 hypocretin Mus musculus 0-4 27071101-3 2016 OX-A is crucial for the control of wakefulness and energy homeostasis and promotes, in OX-1R-expressing cells, the biosynthesis of the endogenous counterpart of marijuana"s psychotropic and appetite-inducing component Delta(9)-tetrahydrocannabinol, i.e., the endocannabinoid 2-arachidonoylglycerol (2-AG), which acts at CB1R. glyceryl 2-arachidonate 299-303 hypocretin Mus musculus 0-4 27071101-4 2016 We report that OX-A/OX-1R signaling at POMC neurons promotes 2-AG biosynthesis, hyperphagia, and weight gain by blunting alpha-MSH production via CB1R-induced and extracellular-signal-regulated kinase 1/2 activation- and STAT3 inhibition-mediated suppression of Pomc gene transcription. glyceryl 2-arachidonate 61-65 hypocretin Mus musculus 15-19 26799708-0 2016 Involvement of the orexin/hypocretin system in the pharmacological effects induced by Delta(9) -tetrahydrocannabinol. Dronabinol 86-116 hypocretin Mus musculus 19-25 26851547-0 2016 The highly selective orexin/hypocretin 1 receptor antagonist GSK1059865 potently reduces ethanol drinking in ethanol dependent mice. 5-bromo-N-(1-(3-fluoro-2-methoxybenzoyl)-5-methylpiperidin-2-yl)methylpyridin-2-amine 61-71 hypocretin Mus musculus 21-27 26851547-0 2016 The highly selective orexin/hypocretin 1 receptor antagonist GSK1059865 potently reduces ethanol drinking in ethanol dependent mice. 5-bromo-N-(1-(3-fluoro-2-methoxybenzoyl)-5-methylpiperidin-2-yl)methylpyridin-2-amine 61-71 hypocretin Mus musculus 28-38 26851547-0 2016 The highly selective orexin/hypocretin 1 receptor antagonist GSK1059865 potently reduces ethanol drinking in ethanol dependent mice. Ethanol 89-96 hypocretin Mus musculus 21-27 26851547-0 2016 The highly selective orexin/hypocretin 1 receptor antagonist GSK1059865 potently reduces ethanol drinking in ethanol dependent mice. Ethanol 89-96 hypocretin Mus musculus 28-38 26851547-0 2016 The highly selective orexin/hypocretin 1 receptor antagonist GSK1059865 potently reduces ethanol drinking in ethanol dependent mice. Ethanol 109-116 hypocretin Mus musculus 21-27 26851547-0 2016 The highly selective orexin/hypocretin 1 receptor antagonist GSK1059865 potently reduces ethanol drinking in ethanol dependent mice. Ethanol 109-116 hypocretin Mus musculus 28-38 26446112-7 2016 Orexin levels in the lateral hypothalamus/perifornical region (LH/P) and hypothalamic pathology were assessed with immunohistochemistry and oxygen polarography. Oxygen 140-146 hypocretin Mus musculus 0-6 27091533-9 2016 Co-administration of modafinil/flecainide resulted in a marked decrease in the number and duration of direct transitions to rapid eye movement sleep, which are characteristic of narcoleptic episodes in orexin knockout mice. Modafinil 21-30 hypocretin Mus musculus 202-208 26492471-0 2016 Nighttime Administration of Nicotine Improves Hepatic Glucose Metabolism via the Hypothalamic Orexin System in Mice. Nicotine 28-36 hypocretin Mus musculus 94-100 26492471-3 2016 The oral intake of nicotine in drinking water, which mainly occurred during the nighttime active period, enhanced daily hypothalamic prepro-orexin gene expression and reduced hyperglycemia in type 2 diabetic db/db mice without affecting body weight, body fat content, and serum levels of insulin. Nicotine 19-27 hypocretin Mus musculus 133-146 26492471-3 2016 The oral intake of nicotine in drinking water, which mainly occurred during the nighttime active period, enhanced daily hypothalamic prepro-orexin gene expression and reduced hyperglycemia in type 2 diabetic db/db mice without affecting body weight, body fat content, and serum levels of insulin. Drinking Water 31-45 hypocretin Mus musculus 133-146 26492471-6 2016 In the pyruvate tolerance test to evaluate hepatic gluconeogenic activity, the oral nicotine treatment moderately suppressed glucose elevations in normal mice and mice lacking dopamine receptors, whereas this effect was abolished in orexin-deficient mice and hepatic parasympathectomized mice. Nicotine 84-92 hypocretin Mus musculus 233-239 26492471-8 2016 These results indicated that the chronic daily administration of nicotine suppressed hepatic gluconeogenesis via the hypothalamic orexin-parasympathetic nervous system. Nicotine 65-73 hypocretin Mus musculus 130-136 26306651-0 2015 Role of orexin A signaling in dietary palmitic acid-activated microglial cells. Palmitic Acid 38-51 hypocretin Mus musculus 8-16 26474479-0 2015 Histamine Transmission Modulates the Phenotype of Murine Narcolepsy Caused by Orexin Neuron Deficiency. Histamine 0-9 hypocretin Mus musculus 78-84 26306651-9 2015 Pro-inflammatory markers IL-6, TNF-alpha, and inducible nitric oxide synthase in microglial cells are increased following PA exposure, but are reduced by pretreatment with OXA. Palmitic Acid 122-124 hypocretin Mus musculus 172-175 26306651-11 2015 Finally, we show hypothalamic neurons exposed to conditioned media from PA-challenged microglia have increased cell survival only when microglia were pretreated with OXA. Palmitic Acid 72-74 hypocretin Mus musculus 166-169 25623402-0 2015 Binge-like consumption of ethanol and other salient reinforcers is blocked by orexin-1 receptor inhibition and leads to a reduction of hypothalamic orexin immunoreactivity. Ethanol 26-33 hypocretin Mus musculus 78-84 26343602-0 2015 7,8-Dihydroxyflavone reduces sleep during dark phase and suppresses orexin A but not orexin B in mice. 6,7-dihydroxyflavone 0-20 hypocretin Mus musculus 68-76 26343602-11 2015 Interestingly, hypothalamic levels of orexin A were also significantly decreased in the DHF group (97 pg/mg) when compared with the vehicle-treated group (132 pg/mg). 6,7-dihydroxyflavone 88-91 hypocretin Mus musculus 38-46 25813215-0 2015 Orexin A regulates plasma insulin and leptin levels in a time-dependent manner following a glucose load in mice. Glucose 91-98 hypocretin Mus musculus 0-8 24297328-0 2015 Hypocretin (orexin) regulates glutamate input to fast-spiking interneurons in layer V of the Fr2 region of the murine prefrontal cortex. Glutamic Acid 30-39 hypocretin Mus musculus 0-10 24297328-6 2015 Finally, hypocretin increased the firing frequency in FS cells, and the effect was blocked when the ionotropic glutamate transmission was inhibited. Glutamic Acid 111-120 hypocretin Mus musculus 9-19 24297328-10 2015 We conclude that 1) hypocretin regulates glutamate release in Fr2; 2) the effect presents a presynaptic component; 3) the peptide control of FS cells is indirect, and probably mediated by the regulation of glutamatergic input onto these cells. Glutamic Acid 41-50 hypocretin Mus musculus 20-30 25586545-10 2015 The OX1R antagonist SB334867 (10-6 M) and AKT antagonist PF-04691502 (10-6 M), when used individually or in combination, abolished the effect of orexin A (10-8 M) on BGC-823 cells. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 20-28 hypocretin Mus musculus 145-153 25586545-10 2015 The OX1R antagonist SB334867 (10-6 M) and AKT antagonist PF-04691502 (10-6 M), when used individually or in combination, abolished the effect of orexin A (10-8 M) on BGC-823 cells. 2-amino-8-(4-(2-hydroxyethoxy)cyclohexyl)-6-(6-methoxypyridin-3-yl)-4-methylpyrido(2,3-d)pyrimidin-7(8H)-one 57-68 hypocretin Mus musculus 145-153 25695914-3 2015 Here using optogenetic approaches in mice, we show that neurons that produce hypocretin (Hcrt)/orexin in the lateral hypothalamic area (LHA) regulate corticosterone release and a variety of behaviours and physiological hallmarks of the stress response. Corticosterone 150-164 hypocretin Mus musculus 89-93 25695914-3 2015 Here using optogenetic approaches in mice, we show that neurons that produce hypocretin (Hcrt)/orexin in the lateral hypothalamic area (LHA) regulate corticosterone release and a variety of behaviours and physiological hallmarks of the stress response. Corticosterone 150-164 hypocretin Mus musculus 95-101 25249578-3 2015 Wild-type mice showed that a daily rhythm in blood glucose levels peaked at the awake period; however, the glucose rhythm disappeared in orexin knockout mice despite normal feeding rhythm. Glucose 107-114 hypocretin Mus musculus 137-143 25249578-4 2015 Central administration of orexin A during nighttime awake period acutely elevated blood glucose levels but subsequently lowered daytime glucose levels in normal and diabetic db/db mice. Glucose 88-95 hypocretin Mus musculus 26-34 25249578-4 2015 Central administration of orexin A during nighttime awake period acutely elevated blood glucose levels but subsequently lowered daytime glucose levels in normal and diabetic db/db mice. Glucose 136-143 hypocretin Mus musculus 26-34 25249578-5 2015 The glucose-elevating and -lowering effects of orexin A were suppressed by adrenergic antagonists and hepatic parasympathectomy, respectively. Glucose 4-11 hypocretin Mus musculus 47-55 25249578-9 2015 Collectively, the daily glucose rhythm under control of orexin appears to be important for maintaining ER homeostasis, thereby preventing insulin resistance in the liver. Glucose 24-31 hypocretin Mus musculus 56-62 25462014-1 2015 Hypothalamic orexin-containing neurons are activated by CO2 and contribute to hypercapnic ventilatory activation. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 56-59 hypocretin Mus musculus 13-19 25462014-7 2015 These results correlate with those of our previous work showing that IH-induced respiratory long-term facilitation is dependent on orexin-containing neurons. Ile-His 69-71 hypocretin Mus musculus 131-137 26500842-3 2015 Stimulatory and inhibitory leptin actions on dopamine neurons have been reported, e.g. by indirect actions on orexin neurons or via direct innervation of dopamine neurons in the ventral tegmental area. Dopamine 45-53 hypocretin Mus musculus 110-116 25953512-6 2015 The investigation afforded a potential therapeutic agent, (1R,2S)-2-{[(2,4-dimethylpyrimidin-5-yl)oxy]methyl}-2-(3-fluorophenyl)-N-(5-fluoropyridin-2-yl)cyclopropanecarboxamide (E2006), an orally active, potent orexin antagonist. Lemborexant 178-183 hypocretin Mus musculus 211-217 25623402-0 2015 Binge-like consumption of ethanol and other salient reinforcers is blocked by orexin-1 receptor inhibition and leads to a reduction of hypothalamic orexin immunoreactivity. Ethanol 26-33 hypocretin Mus musculus 148-154 25623402-2 2015 Consistent with these findings, a growing number of studies have implicated the OX system in modulating the rewarding properties of several drugs of abuse, including ethanol (EtOH). Ethanol 166-173 hypocretin Mus musculus 80-82 25623402-2 2015 Consistent with these findings, a growing number of studies have implicated the OX system in modulating the rewarding properties of several drugs of abuse, including ethanol (EtOH). Ethanol 175-179 hypocretin Mus musculus 80-82 25623402-3 2015 However, the role of the OX system in excessive binge-like EtOH intake remains relatively unexplored. Ethanol 59-63 hypocretin Mus musculus 25-27 25623402-4 2015 Here, we assessed changes in OX immunoreactivity (IR) in the hypothalamus following repeated cycles of binge-like EtOH drinking and assessed the participation of the OX-1 receptor (OX1R) in binge-like EtOH consumption. Ethanol 114-118 hypocretin Mus musculus 29-31 25623402-11 2015 CONCLUSIONS: Our observed reduction in OX IR in the LH indicates that the OX system in engaged during binge-like consumption of EtOH and sucrose. Ethanol 128-132 hypocretin Mus musculus 39-41 25623402-11 2015 CONCLUSIONS: Our observed reduction in OX IR in the LH indicates that the OX system in engaged during binge-like consumption of EtOH and sucrose. Sucrose 137-144 hypocretin Mus musculus 39-41 25623402-12 2015 The observation that inhibition of the OX1R signaling blunted binge-like EtOH, and saccharin drinking suggests that reward-related OX circuits originating in the LH participate in the consumption of salient reinforcers regardless of calories. Ethanol 73-77 hypocretin Mus musculus 39-41 25195718-2 2014 We have recently demonstrated that sleep deprivation stimulates local nitric oxide (NO) production by neuronal NO synthase in the lateral hypothalamus, which leads to selective degeneration of orexin neurons accompanied by formation of orexin-immunoreactive aggregates. Nitric Oxide 70-82 hypocretin Mus musculus 193-199 25994341-10 2015 In addition, the mice showed disturbance of orexin neuron activity related to the sleep-arousal system, which is involved in fatigue symptoms under fasting condition, one of the states showing enhanced fatty acid metabolism. Fatty Acids 202-212 hypocretin Mus musculus 44-50 25195718-2 2014 We have recently demonstrated that sleep deprivation stimulates local nitric oxide (NO) production by neuronal NO synthase in the lateral hypothalamus, which leads to selective degeneration of orexin neurons accompanied by formation of orexin-immunoreactive aggregates. Nitric Oxide 70-82 hypocretin Mus musculus 236-242 25278019-0 2014 Orexin/hypocretin activates mTOR complex 1 (mTORC1) via an Erk/Akt-independent and calcium-stimulated lysosome v-ATPase pathway. Calcium 83-90 hypocretin Mus musculus 0-6 25278019-0 2014 Orexin/hypocretin activates mTOR complex 1 (mTORC1) via an Erk/Akt-independent and calcium-stimulated lysosome v-ATPase pathway. Calcium 83-90 hypocretin Mus musculus 7-17 25278019-4 2014 This orexin/GPCR-stimulated mTOR activation is sensitive to rapamycin, an inhibitor of mTOR complex 1 (mTORC1) but is independent of two well known mTORC1 activators, Erk and Akt. Sirolimus 60-69 hypocretin Mus musculus 5-11 25278019-5 2014 Rather, our studies indicate that orexin activates mTORC1 via extracellular calcium influx and the lysosome pathway involving v-ATPase and Rag GTPases. Calcium 76-83 hypocretin Mus musculus 34-40 25278019-6 2014 Moreover, a cytoplasmic calcium transient is sufficient to mimic orexin/GPCR signaling to mTORC1 activation in a v-ATPase-dependent manner. Calcium 24-31 hypocretin Mus musculus 65-71 24930888-6 2014 The extinction-facilitating effects of the hypocretin receptor-1 antagonist SB334867 were associated with increased expression of cFos in the basolateral amygdala and the infralimbic cortex. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 76-84 hypocretin Mus musculus 43-53 24983661-3 2014 The present study evaluates from a pharmacological and a molecular approach the contribution of OX to excessive binge-like consumption of highly preferred palatable substances (sucrose and saccharin) in ad libitum-fed C57BL/6J mice. Sucrose 177-184 hypocretin Mus musculus 96-98 24983661-3 2014 The present study evaluates from a pharmacological and a molecular approach the contribution of OX to excessive binge-like consumption of highly preferred palatable substances (sucrose and saccharin) in ad libitum-fed C57BL/6J mice. Saccharin 189-198 hypocretin Mus musculus 96-98 24983661-5 2014 (2) Four repetitive, 2-h daily episodes of sucrose and saccharin (but not water) binge-like drinking significantly dampened OX mRNA expression in the LH. Sucrose 43-50 hypocretin Mus musculus 124-126 24983661-5 2014 (2) Four repetitive, 2-h daily episodes of sucrose and saccharin (but not water) binge-like drinking significantly dampened OX mRNA expression in the LH. Saccharin 55-64 hypocretin Mus musculus 124-126 24951857-2 2014 Past studies have shown the role of the orexin peptide itself; however, orexin neurons contain not only orexin but also other neurotransmitters such as glutamate and dynorphin. Glutamic Acid 152-161 hypocretin Mus musculus 72-78 24951857-2 2014 Past studies have shown the role of the orexin peptide itself; however, orexin neurons contain not only orexin but also other neurotransmitters such as glutamate and dynorphin. Glutamic Acid 152-161 hypocretin Mus musculus 72-78 24951857-5 2014 By intraperitoneal injection of clozapine-N oxide in orexin-Cre mice expressing hM3Dq in orexin neurons, we could selectively manipulate the activity of orexin neurons. clozapine N-oxide 32-49 hypocretin Mus musculus 53-59 24951857-5 2014 By intraperitoneal injection of clozapine-N oxide in orexin-Cre mice expressing hM3Dq in orexin neurons, we could selectively manipulate the activity of orexin neurons. clozapine N-oxide 32-49 hypocretin Mus musculus 89-95 24951857-5 2014 By intraperitoneal injection of clozapine-N oxide in orexin-Cre mice expressing hM3Dq in orexin neurons, we could selectively manipulate the activity of orexin neurons. clozapine N-oxide 32-49 hypocretin Mus musculus 89-95 24951857-5 2014 By intraperitoneal injection of clozapine-N oxide in orexin-Cre mice expressing hM3Dq in orexin neurons, we could selectively manipulate the activity of orexin neurons. hm3dq 80-85 hypocretin Mus musculus 53-59 24951857-5 2014 By intraperitoneal injection of clozapine-N oxide in orexin-Cre mice expressing hM3Dq in orexin neurons, we could selectively manipulate the activity of orexin neurons. hm3dq 80-85 hypocretin Mus musculus 89-95 24951857-5 2014 By intraperitoneal injection of clozapine-N oxide in orexin-Cre mice expressing hM3Dq in orexin neurons, we could selectively manipulate the activity of orexin neurons. hm3dq 80-85 hypocretin Mus musculus 89-95 24951857-6 2014 Pharmacogenetic stimulation of orexin neurons simultaneously increased locomotive activity, food intake, water intake and the respiratory exchange ratio (RER). Water 105-110 hypocretin Mus musculus 31-37 24951857-8 2014 Accordantly, 83% ablation of orexin neurons resulted in decreased food and water intake, while 70% ablation had almost no effect on these parameters. Water 75-80 hypocretin Mus musculus 29-35 24806676-4 2014 Upon doxycycline removal from the diet of postpubertal orexin-tTA;TetO DTA mice, orexin neurodegeneration was rapid, with 80% cell loss within 7 d, and resulted in disrupted sleep architecture. Doxycycline 5-16 hypocretin Mus musculus 55-61 25107627-0 2014 Metabolic regulation of lateral hypothalamic glucose-inhibited orexin neurons may influence midbrain reward neurocircuitry. Glucose 45-52 hypocretin Mus musculus 63-69 25107627-1 2014 Lateral hypothalamic area (LHA) orexin neurons modulate reward-based feeding by activating ventral tegmental area (VTA) dopamine (DA) neurons. Dopamine 120-128 hypocretin Mus musculus 32-38 25107627-1 2014 Lateral hypothalamic area (LHA) orexin neurons modulate reward-based feeding by activating ventral tegmental area (VTA) dopamine (DA) neurons. Dopamine 130-132 hypocretin Mus musculus 32-38 25107627-2 2014 We hypothesize that signals of peripheral energy status influence reward-based feeding by modulating the glucose sensitivity of LHA orexin glucose-inhibited (GI) neurons. Glucose 105-112 hypocretin Mus musculus 132-138 25107627-4 2014 Low glucose directly activated ~60% of LHA orexin-GFP neurons in both whole cell and cell attached recordings. Glucose 4-11 hypocretin Mus musculus 43-49 25107627-5 2014 Leptin indirectly reduced and ghrelin directly enhanced the activation of LHA orexin-GI neurons by glucose decreases from 2.5 to 0.1mM by 53+-12% (n=16, P<0.001) and 41+-24% (n=8, P<0.05), respectively. Glucose 99-106 hypocretin Mus musculus 78-84 25107627-7 2014 Fasting increased activation of LHA orexin-GI neurons by decreased glucose, as would be predicted by these hormonal effects. Glucose 67-74 hypocretin Mus musculus 36-42 25107627-11 2014 Orexin-1 but not 2 receptor antagonism with SB334867 (10muM; n=9) and TCS-OX2-29 (2muM; n=5), respectively, blocks the effects of decreased glucose on VTA DA neurons. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 44-52 hypocretin Mus musculus 0-6 25107627-11 2014 Orexin-1 but not 2 receptor antagonism with SB334867 (10muM; n=9) and TCS-OX2-29 (2muM; n=5), respectively, blocks the effects of decreased glucose on VTA DA neurons. Glucose 140-147 hypocretin Mus musculus 0-6 25107627-12 2014 Thus, decreased glucose increases orexin-dependent excitatory glutamate neurotransmission onto VTA DA neurons. Glucose 16-23 hypocretin Mus musculus 34-40 25107627-12 2014 Thus, decreased glucose increases orexin-dependent excitatory glutamate neurotransmission onto VTA DA neurons. Glutamic Acid 62-71 hypocretin Mus musculus 34-40 25107627-13 2014 These data suggest that the glucose sensitivity of LHA orexin-GI neurons links metabolic state and reward-based feeding. Glucose 28-35 hypocretin Mus musculus 55-61 25143620-0 2014 Leptin acts via lateral hypothalamic area neurotensin neurons to inhibit orexin neurons by multiple GABA-independent mechanisms. gamma-Aminobutyric Acid 100-104 hypocretin Mus musculus 73-79 24874709-0 2014 Orexin A-induced anxiety-like behavior is mediated through GABA-ergic, alpha- and beta-adrenergic neurotransmissions in mice. gamma-Aminobutyric Acid 59-63 hypocretin Mus musculus 0-8 24874709-8 2014 Our results show that orexin A decreases significantly the time spent in the arms (open/open+closed) and this action is reversed by bicuculline, phenoxybenzamine and propranolol, but not by atropine, haloperidol or nitro-l-arginine. Bicuculline 132-143 hypocretin Mus musculus 22-30 24874709-8 2014 Our results show that orexin A decreases significantly the time spent in the arms (open/open+closed) and this action is reversed by bicuculline, phenoxybenzamine and propranolol, but not by atropine, haloperidol or nitro-l-arginine. Phenoxybenzamine 145-161 hypocretin Mus musculus 22-30 24874709-8 2014 Our results show that orexin A decreases significantly the time spent in the arms (open/open+closed) and this action is reversed by bicuculline, phenoxybenzamine and propranolol, but not by atropine, haloperidol or nitro-l-arginine. Propranolol 166-177 hypocretin Mus musculus 22-30 24874709-8 2014 Our results show that orexin A decreases significantly the time spent in the arms (open/open+closed) and this action is reversed by bicuculline, phenoxybenzamine and propranolol, but not by atropine, haloperidol or nitro-l-arginine. Nitroarginine 215-231 hypocretin Mus musculus 22-30 24874709-9 2014 Our results provide evidence for the first time that the orexin A-induced anxiety-like behavior is mediated through GABA-A-ergic, alpha- and beta-adrenergic neurotransmissions, whereas muscarinic cholinergic, dopaminergic and nitrergic neurotransmissions may not be implicated. gamma-Aminobutyric Acid 116-120 hypocretin Mus musculus 57-65 24806676-4 2014 Upon doxycycline removal from the diet of postpubertal orexin-tTA;TetO DTA mice, orexin neurodegeneration was rapid, with 80% cell loss within 7 d, and resulted in disrupted sleep architecture. Doxycycline 5-16 hypocretin Mus musculus 81-87 24478100-7 2014 Using Neuro-2a cells, a mouse neuroblastoma cell line, TeNT HCR (HCR/T) and TeNT(RY) were found to bind gangliosides with similar affinities and specificities, consistent with the HCR domain containing receptor binding function. Gangliosides 104-116 hypocretin Mus musculus 60-63 24806676-4 2014 Upon doxycycline removal from the diet of postpubertal orexin-tTA;TetO DTA mice, orexin neurodegeneration was rapid, with 80% cell loss within 7 d, and resulted in disrupted sleep architecture. 1-(carboxymethylthio)tetradecane 61-65 hypocretin Mus musculus 81-87 24806676-7 2014 Temporary doxycycline removal followed by reintroduction after several days enabled partial lesion of orexin neurons. Doxycycline 10-21 hypocretin Mus musculus 102-108 24790280-3 2014 In the PF-LHA, HCRT neurons are intermingled with glutamate-expressing neurons and also co-express glutamate. Glutamic Acid 99-108 hypocretin Mus musculus 15-19 24790280-4 2014 Evidence suggests that HCRT-glutamate interactions within the PF-LHA may play a critical role in maintaining behavioral arousal. Glutamic Acid 28-37 hypocretin Mus musculus 23-27 24790280-7 2014 We used reverse microdialysis to deliver N-methyl-D-aspartate (NMDA) into the HCRT zone of the PF-LHA in HCRT-KO and wild-type (WT) mice to evaluate the contributions of glutamatergic vs. HCRT signaling in sleep-wake regulation. N-Methylaspartate 63-67 hypocretin Mus musculus 78-82 24790280-8 2014 MEASUREMENTS AND RESULTS: As compared to respective controls, local perfusion of NMDA into the PF-LHA, dose-dependently increased active-waking with concomitant reductions in nonREM and REM sleep in spontaneously sleeping WT as well as HCRT-KO mice. N-Methylaspartate 81-85 hypocretin Mus musculus 236-240 24790280-9 2014 However, compared to WT, the NMDA-induced behavioral changes in HCRT-KO mice were significantly attenuated, as evidenced by the higher dose of NMDA needed and lower magnitude of changes induced in sleep-wake parameters. N-Methylaspartate 29-33 hypocretin Mus musculus 64-68 24790280-9 2014 However, compared to WT, the NMDA-induced behavioral changes in HCRT-KO mice were significantly attenuated, as evidenced by the higher dose of NMDA needed and lower magnitude of changes induced in sleep-wake parameters. N-Methylaspartate 143-147 hypocretin Mus musculus 64-68 24790280-10 2014 Although not observed in WT mice, the number of cataplectic events increased significantly during NMDA-induced behavioral arousal in HCRT-KO mice. N-Methylaspartate 98-102 hypocretin Mus musculus 133-137 24368186-7 2014 In addition, the cAMP response element reporter activities were significantly reduced, whereas the serum response element luciferase and the T-lymphocyte activation of nuclear factor-responsive element reporter activity were significantly up-regulated after Orexin(s) stimulation. Cyclic AMP 17-21 hypocretin Mus musculus 258-264 24759941-1 2014 Orexin-A (a neuropeptide in the hypothalamus) plays an important role in many physiological functions, including the regulation of glucose metabolism. Glucose 131-138 hypocretin Mus musculus 0-8 24478100-7 2014 Using Neuro-2a cells, a mouse neuroblastoma cell line, TeNT HCR (HCR/T) and TeNT(RY) were found to bind gangliosides with similar affinities and specificities, consistent with the HCR domain containing receptor binding function. Gangliosides 104-116 hypocretin Mus musculus 65-70 24478100-7 2014 Using Neuro-2a cells, a mouse neuroblastoma cell line, TeNT HCR (HCR/T) and TeNT(RY) were found to bind gangliosides with similar affinities and specificities, consistent with the HCR domain containing receptor binding function. Gangliosides 104-116 hypocretin Mus musculus 65-68 24466352-4 2014 We also found that: i- HFD-mediated orosensory stimulation was required for the mesolimbic pathway activation, ii- acute HFD differentially activates dopamine neurons of the paranigral, parabrachial pigmented and interfascicular sub-regions of the VTA, and iii- orexin neurons of the lateral hypothalamic area are responsive to acute HFD. Dopamine 150-158 hypocretin Mus musculus 262-268 24466352-5 2014 Moreover, orexin signaling blockade, with the orexin 1 receptor antagonist SB-334867, reduces acute HFD consumption and c-Fos induction in the VTA but not in the other mesolimbic nuclei under study. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 75-84 hypocretin Mus musculus 10-16 24466352-5 2014 Moreover, orexin signaling blockade, with the orexin 1 receptor antagonist SB-334867, reduces acute HFD consumption and c-Fos induction in the VTA but not in the other mesolimbic nuclei under study. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 75-84 hypocretin Mus musculus 46-52 23707248-5 2013 We further confirmed that green light illumination for 1h in the dark period suppressed orexin neuronal activity in vivo using c-Fos expression. Hydrogen 55-57 hypocretin Mus musculus 88-94 24574958-6 2014 The prepro-orexin knockout mouse has a significantly attenuated ventilatory CO2 chemoreflex, and in normal rats, central application of orexin stimulates breathing while blocking orexin receptors decreases the ventilatory CO2 chemoreflex. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 76-79 hypocretin Mus musculus 11-17 24574958-6 2014 The prepro-orexin knockout mouse has a significantly attenuated ventilatory CO2 chemoreflex, and in normal rats, central application of orexin stimulates breathing while blocking orexin receptors decreases the ventilatory CO2 chemoreflex. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 222-225 hypocretin Mus musculus 11-17 24574958-6 2014 The prepro-orexin knockout mouse has a significantly attenuated ventilatory CO2 chemoreflex, and in normal rats, central application of orexin stimulates breathing while blocking orexin receptors decreases the ventilatory CO2 chemoreflex. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 222-225 hypocretin Mus musculus 136-142 24574958-7 2014 Interestingly, SHRs have a significantly increased ventilatory CO2 chemoreflex relative to normotensive WKY rats and blocking both orexin receptors can normalize this exaggerated response. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 63-66 hypocretin Mus musculus 131-137 24574958-10 2014 Treating SHRs by blocking both orexin receptors with oral administration of an antagonist, almorexant (Almxt), can normalize the CO2 chemoreflex and significantly lower ABP and SNA. almorexant 91-101 hypocretin Mus musculus 31-37 24574958-10 2014 Treating SHRs by blocking both orexin receptors with oral administration of an antagonist, almorexant (Almxt), can normalize the CO2 chemoreflex and significantly lower ABP and SNA. almorexant 103-108 hypocretin Mus musculus 31-37 24574958-10 2014 Treating SHRs by blocking both orexin receptors with oral administration of an antagonist, almorexant (Almxt), can normalize the CO2 chemoreflex and significantly lower ABP and SNA. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 129-132 hypocretin Mus musculus 31-37 24391530-7 2013 Considering published behavioral data, these findings support a model in which orexin-mediated excitation of mesopontine cholinergic and monoaminergic neurons contributes little to stabilizing spontaneous waking and sleep bouts, but functions in context-dependent arousal and helps restrict muscle atonia to REM sleep. mesopontine 109-120 hypocretin Mus musculus 79-85 23959674-0 2013 Orexin neurons are indispensable for prostaglandin E2-induced fever and defence against environmental cooling in mice. Dinoprostone 37-53 hypocretin Mus musculus 0-6 23959674-6 2013 In WT and ORX-KO mice, the administration of PGE2 and cold exposure activated orexin neurons, as revealed by increased levels of expression of c-fos. Dinoprostone 45-49 hypocretin Mus musculus 78-84 23959674-10 2013 In addition, these results indicate the possible involvement of glutamate in orexin neurons implicated in PGE2-induced fever. Glutamic Acid 64-73 hypocretin Mus musculus 77-83 23959674-10 2013 In addition, these results indicate the possible involvement of glutamate in orexin neurons implicated in PGE2-induced fever. Dinoprostone 106-110 hypocretin Mus musculus 77-83 24223949-7 2013 Twelve genes, including Ptgds, Hcrt, Tmed2, Klc1, and Nedd4, whose mRNA expressions were down-regulated by the neonatal EB treatment, were chosen for further examination by semiquantitative RT-PCR in the hypothalamus of perinatal intact male and female mice. estradiol 3-benzoate 120-122 hypocretin Mus musculus 31-35 24223949-10 2013 Some genes, such as Ptgds encoding prostaglandin D2 production enzyme and Hcrt encording orexin, have been reported to have a role in neuroprotection. Prostaglandin D2 35-51 hypocretin Mus musculus 89-95 23707248-6 2013 Continuous 1h silencing of orexin neurons in freely moving orexin-tTA; TetO ArchT mice during the night (the active period, 20:00-21:00) significantly increased total time spent in slow-wave sleep (SWS) and decreased total wake time. Hydrogen 11-13 hypocretin Mus musculus 27-33 23707248-6 2013 Continuous 1h silencing of orexin neurons in freely moving orexin-tTA; TetO ArchT mice during the night (the active period, 20:00-21:00) significantly increased total time spent in slow-wave sleep (SWS) and decreased total wake time. Hydrogen 11-13 hypocretin Mus musculus 59-65 23880022-2 2013 In the present study we evaluated the role of orexin in ethanol-induced behavioral sensitization. Ethanol 56-63 hypocretin Mus musculus 46-52 23880022-7 2013 In the second experiment, mice were treated as in Experiment 1 and type 1 orexin receptor antagonist, SB334867 (20mg/kg), was administered before the ethanol challenge successfully blocking the expression of sensitization in mice chronically treated with EtOH. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 102-110 hypocretin Mus musculus 74-80 23880022-8 2013 These results indicate that orexin plays a role in ethanol-induced behavioral sensitization. Ethanol 51-58 hypocretin Mus musculus 28-34 23408466-8 2013 Risperidone induced a higher UCP1 mRNA (P = 0.003) and a lower orexin mRNA (P = 0.001) than placebo. Risperidone 0-11 hypocretin Mus musculus 63-69 23997373-3 2013 DESIGN: 5HT1A receptor expression level was specifically and reversibly controlled in the orexin neurons using the Tet-off system. tetramethylenedisulfotetramine 115-118 hypocretin Mus musculus 90-96 23997373-4 2013 The responsiveness of orexin neurons to 5HT in vitro and the sleep/wakefulness patterns were compared between 5HT1A-overexpressing and control mice. Serotonin 40-43 hypocretin Mus musculus 22-28 23997373-5 2013 MEASUREMENTS AND RESULTS: When the 5HT1A receptor was overexpressed in orexin neurons of Orexin-EGFP; orexin-tTA; TetO Htr1a mice, 5HT-induced inhibition of orexin neurons was prolonged. Serotonin 35-38 hypocretin Mus musculus 71-77 23997373-5 2013 MEASUREMENTS AND RESULTS: When the 5HT1A receptor was overexpressed in orexin neurons of Orexin-EGFP; orexin-tTA; TetO Htr1a mice, 5HT-induced inhibition of orexin neurons was prolonged. Serotonin 35-38 hypocretin Mus musculus 89-95 23997373-5 2013 MEASUREMENTS AND RESULTS: When the 5HT1A receptor was overexpressed in orexin neurons of Orexin-EGFP; orexin-tTA; TetO Htr1a mice, 5HT-induced inhibition of orexin neurons was prolonged. Serotonin 35-38 hypocretin Mus musculus 102-108 23997373-5 2013 MEASUREMENTS AND RESULTS: When the 5HT1A receptor was overexpressed in orexin neurons of Orexin-EGFP; orexin-tTA; TetO Htr1a mice, 5HT-induced inhibition of orexin neurons was prolonged. Serotonin 35-38 hypocretin Mus musculus 102-108 23997373-7 2013 However, when the 5HT1A receptor in orexin neurons was reduced to basal expression levels in the presence of doxycycline, sleep/wakefulness patterns in Orexin-tTA; TetO Htr1a mice during the early active period were indistinguishable from those of littermate TetO Htr1a mice. Doxycycline 109-120 hypocretin Mus musculus 36-42 23904594-4 2013 An endoplasmic reticulum stress inducer tunicamycin replicated the effect of NOC18 with regard to decrease of orexin-IR neurons and formation of aggregates. Tunicamycin 40-51 hypocretin Mus musculus 110-116 23904594-6 2013 Moreover, PDI inhibitors, such as cystamine and securinine, caused a selective decrease of orexin neurons and promoted formation of orexin-A-IR aggregates. Cystamine 34-43 hypocretin Mus musculus 91-97 23904594-6 2013 Moreover, PDI inhibitors, such as cystamine and securinine, caused a selective decrease of orexin neurons and promoted formation of orexin-A-IR aggregates. Cystamine 34-43 hypocretin Mus musculus 132-138 23904594-6 2013 Moreover, PDI inhibitors, such as cystamine and securinine, caused a selective decrease of orexin neurons and promoted formation of orexin-A-IR aggregates. securinine 48-58 hypocretin Mus musculus 91-97 23904594-6 2013 Moreover, PDI inhibitors, such as cystamine and securinine, caused a selective decrease of orexin neurons and promoted formation of orexin-A-IR aggregates. securinine 48-58 hypocretin Mus musculus 132-138 23625921-4 2013 Considering that the metabolic status affects orexin expression, we supplemented the culture medium with a nutrient factor, ManNAc, and succeeded in generating functional orexin neurons from mouse ES cells. N-acetylmannosamine 124-130 hypocretin Mus musculus 46-52 23904594-0 2013 Nitric oxide mediates selective degeneration of hypothalamic orexin neurons through dysfunction of protein disulfide isomerase. Nitric Oxide 0-12 hypocretin Mus musculus 61-67 23904594-1 2013 We addressed the role of nitric oxide (NO) in orexin neuron degeneration that has been observed under various pathological conditions. Nitric Oxide 25-37 hypocretin Mus musculus 46-52 23625921-4 2013 Considering that the metabolic status affects orexin expression, we supplemented the culture medium with a nutrient factor, ManNAc, and succeeded in generating functional orexin neurons from mouse ES cells. N-acetylmannosamine 124-130 hypocretin Mus musculus 171-177 23625921-7 2013 In the orexin neurons induced by supplementation of ManNAc, the T-DMR of the Hcrt gene was hypomethylated in association with higher H3/H4 acetylation. N-acetylmannosamine 52-58 hypocretin Mus musculus 7-13 23625921-7 2013 In the orexin neurons induced by supplementation of ManNAc, the T-DMR of the Hcrt gene was hypomethylated in association with higher H3/H4 acetylation. N-acetylmannosamine 52-58 hypocretin Mus musculus 77-81 23625921-9 2013 In non-orexin-expressing cells, H3/H4 hypoacetylation and hyper-O-GlcNAc modification were observed at the T-DMRs occupied by O-GlcNAc transferase and Sirt1. hyper-o-glcnac 58-72 hypocretin Mus musculus 7-13 23625921-9 2013 In non-orexin-expressing cells, H3/H4 hypoacetylation and hyper-O-GlcNAc modification were observed at the T-DMRs occupied by O-GlcNAc transferase and Sirt1. t-dmrs 107-113 hypocretin Mus musculus 7-13 23625921-10 2013 Therefore, the results of the present study suggest that the glucose metabolite, ManNAc, induces switching from the inactive state by Ogt-Sirt1 to the active state by Mgea5, p300, and CBP at the Hcrt gene locus. Glucose 61-68 hypocretin Mus musculus 195-199 23625921-10 2013 Therefore, the results of the present study suggest that the glucose metabolite, ManNAc, induces switching from the inactive state by Ogt-Sirt1 to the active state by Mgea5, p300, and CBP at the Hcrt gene locus. N-acetylmannosamine 81-87 hypocretin Mus musculus 195-199 23373812-8 2013 RSG administration rescued brain derived neurotrophic factor(BDNF) deficiency in the cerebral cortex, and prevented loss of orexin-A-immunopositive neurons in the hypothalamus of N171-82Q HD mice. Rosiglitazone 0-3 hypocretin Mus musculus 124-132 23510906-5 2013 Moreover, orexin-deficient mice maintained under ad libitum-fed conditions after defeat stress exhibited hyperinsulinemia and elevated HOMA-IR (homeostasis model assessment for insulin resistance), despite normal fasting blood glucose levels. Glucose 227-234 hypocretin Mus musculus 10-16 23510906-6 2013 In a pyruvate tolerance test to evaluate hepatic insulin sensitivity, chronic stress-induced abnormal glucose elevation was observed in orexin-deficient but not wild-type mice, although both types of mice were susceptible to chronic stress. Pyruvic Acid 5-13 hypocretin Mus musculus 136-142 23510906-6 2013 In a pyruvate tolerance test to evaluate hepatic insulin sensitivity, chronic stress-induced abnormal glucose elevation was observed in orexin-deficient but not wild-type mice, although both types of mice were susceptible to chronic stress. Glucose 102-109 hypocretin Mus musculus 136-142 23477964-0 2013 The impact of hypothermia on emergence from isoflurane anesthesia in orexin neuron-ablated mice. Isoflurane 44-54 hypocretin Mus musculus 69-75 23516292-12 2013 Our studies demonstrate that Hcrt neurons play an important role in the consolidation of social recognition memory, at least in part through enhancements of hippocampal synaptic plasticity and cAMP response element-binding protein phosphorylation. Cyclic AMP 193-197 hypocretin Mus musculus 29-33 23318871-0 2013 Repeated in vivo exposure of cocaine induces long-lasting synaptic plasticity in hypocretin/orexin-producing neurons in the lateral hypothalamus in mice. Cocaine 29-36 hypocretin Mus musculus 92-98 23255725-10 2013 Hypocretin excited MCH cells by activating a sodium-calcium exchanger and by reducing potassium currents; NPY reduced activity by increasing an inwardly rectifying potassium current. Potassium 86-95 hypocretin Mus musculus 0-10 23508038-6 2013 For example, orexin neurons are regulated by peripheral metabolic cues, including ghrelin, leptin, and glucose concentration. Glucose 103-110 hypocretin Mus musculus 13-19 22995645-3 2013 We investigated the effects of systemic administration of orexin-1-receptor antagonist, SB 334867, and orexin-2 receptor antagonist, TCS-OX2-29 on the acquisition and expression of morphine conditioned place preference (CPP) in both naive and morphine-dependent mice. Morphine 181-189 hypocretin Mus musculus 103-109 22995645-7 2013 The rewarding effect of morphine has stronger correlation with orexin-2 receptors in morphine-dependent mice while it depends on both kinds of receptors in naive mice. Morphine 24-32 hypocretin Mus musculus 63-69 22995645-7 2013 The rewarding effect of morphine has stronger correlation with orexin-2 receptors in morphine-dependent mice while it depends on both kinds of receptors in naive mice. Morphine 85-93 hypocretin Mus musculus 63-69 23117790-1 2013 Orexin-A (a glucose-sensing neuropeptide in the hypothalamus) and brain-derived neurotrophic factor (BDNF; a member of the neurotrophin family) play roles in many physiologic functions, including regulation of glucose metabolism. Glucose 12-19 hypocretin Mus musculus 0-8 23117790-1 2013 Orexin-A (a glucose-sensing neuropeptide in the hypothalamus) and brain-derived neurotrophic factor (BDNF; a member of the neurotrophin family) play roles in many physiologic functions, including regulation of glucose metabolism. Glucose 210-217 hypocretin Mus musculus 0-8 23117790-11 2013 These results suggest that suppression of postischemic glucose intolerance by orexin-A assists in the prevention of cerebral ischemic neuronal damage. Glucose 55-62 hypocretin Mus musculus 78-86 23117790-9 2013 Intrahypothalamic administration of orexin-A (1 or 5 pmol/mouse) significantly and dose-dependently suppressed the development of postischemic glucose intolerance on day 1 and development of neuronal damage on day 3. Glucose 143-150 hypocretin Mus musculus 36-44 23117790-10 2013 The MCAO-induced decrease in insulin receptor levels in the liver and skeletal muscle on day 1 was recovered to control levels by orexin-A, and this effect of orexin-A was reversed by the administration of SB334867 as well as by hypothalamic BDNF knockdown. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 206-214 hypocretin Mus musculus 159-167 22956835-3 2012 A major pathway by which hcrt/orx neurons are thought to promote arousal is through projections to tuberomammillary histamine (HA) neurons. Histamine 116-125 hypocretin Mus musculus 25-29 23204605-0 2012 The dual orexin receptor antagonist almorexant induces sleep and decreases orexin-induced locomotion by blocking orexin 2 receptors. almorexant 36-46 hypocretin Mus musculus 9-15 23204605-0 2012 The dual orexin receptor antagonist almorexant induces sleep and decreases orexin-induced locomotion by blocking orexin 2 receptors. almorexant 36-46 hypocretin Mus musculus 75-81 23204605-0 2012 The dual orexin receptor antagonist almorexant induces sleep and decreases orexin-induced locomotion by blocking orexin 2 receptors. almorexant 36-46 hypocretin Mus musculus 75-81 22956835-3 2012 A major pathway by which hcrt/orx neurons are thought to promote arousal is through projections to tuberomammillary histamine (HA) neurons. Histamine 127-129 hypocretin Mus musculus 25-29 22453138-0 2012 Activation of the orexin 1 receptor is a critical component of CO2-mediated anxiety and hypertension but not bradycardia. N2,N6-bis(4-(2-aminoethoxy)quinolin-2-yl)-4-((4-fluorobenzyl)oxy)pyridine-2,6-dicarboxamide 63-66 hypocretin Mus musculus 18-24 22652455-14 2012 Orexin B induced ERK phosphorylation in mouse hypothalamus neuron cells differs from CHO cell line and cannot be inhibited by PKC inhibitor GF 109203X. bisindolylmaleimide I 140-150 hypocretin Mus musculus 0-8 22617356-0 2012 Yohimbine depresses excitatory transmission in BNST and impairs extinction of cocaine place preference through orexin-dependent, norepinephrine-independent processes. Yohimbine 0-9 hypocretin Mus musculus 111-117 22617356-3 2012 Recent studies show yohimbine-induced drug-seeking behavior is attenuated by orexin receptor 1 (OX(1)R) antagonists. Yohimbine 20-29 hypocretin Mus musculus 77-83 22617356-5 2012 Here, we investigated yohimbine-orexin interactions. Yohimbine 22-31 hypocretin Mus musculus 32-38 22617356-6 2012 Our results demonstrate yohimbine-induced depression of excitatory transmission in the BNST is unaffected by alpha1-AR and corticotropin-releasing factor receptor-1 (CRFR(1)) antagonists, but is (1) blocked by OxR antagonists and (2) absent in brain slices from orexin knockout mice. Yohimbine 24-33 hypocretin Mus musculus 262-268 22617356-7 2012 Although the actions of yohimbine were not mimicked by the norepinephrine transporter blocker reboxetine, they were by exogenously applied orexin A. Yohimbine 24-33 hypocretin Mus musculus 139-147 22617356-8 2012 We find that, as with yohimbine, orexin A depression of excitatory transmission in BNST is OX(1)R-dependent. Yohimbine 22-31 hypocretin Mus musculus 33-41 22617356-10 2012 These data highlight a new mechanism for orexin on excitatory anxiety circuits and demonstrate that some of the actions of yohimbine may be directly dependent upon orexin signaling and independent of norepinephrine and CRF in the BNST. Yohimbine 123-132 hypocretin Mus musculus 41-47 22617356-10 2012 These data highlight a new mechanism for orexin on excitatory anxiety circuits and demonstrate that some of the actions of yohimbine may be directly dependent upon orexin signaling and independent of norepinephrine and CRF in the BNST. Yohimbine 123-132 hypocretin Mus musculus 164-170 22641088-0 2012 Ghrelin-deficient mice have fewer orexin cells and reduced cFOS expression in the mesolimbic dopamine pathway under a restricted feeding paradigm. Ghrelin 0-7 hypocretin Mus musculus 34-40 22453138-3 2012 Recent evidence has demonstrated that wake-promoting hypothalamic orexin (ORX: also known as hypocretin) neurons are highly sensitive to local changes in CO(2)/H(+), and mice lacking prepro-ORX have blunted respiratory responses to hypercapnia. co(2) 154-159 hypocretin Mus musculus 66-72 22453138-3 2012 Recent evidence has demonstrated that wake-promoting hypothalamic orexin (ORX: also known as hypocretin) neurons are highly sensitive to local changes in CO(2)/H(+), and mice lacking prepro-ORX have blunted respiratory responses to hypercapnia. co(2) 154-159 hypocretin Mus musculus 93-103 21911618-6 2011 In the duodenal preparations, OXA (0.3 muM) caused a TTX-insensitive transient contraction. Tetrodotoxin 53-56 hypocretin Mus musculus 30-33 22234465-8 2012 The reduction of Npy and orexin in hypothalamic cultures was completely prevented by candesartan or the AMPK activator 5-aminoimidazole-4-carboxamide ribonucleoside. candesartan 85-96 hypocretin Mus musculus 25-31 22234465-8 2012 The reduction of Npy and orexin in hypothalamic cultures was completely prevented by candesartan or the AMPK activator 5-aminoimidazole-4-carboxamide ribonucleoside. acadesine 119-164 hypocretin Mus musculus 25-31 21831361-8 2012 Nicotine withdrawal increased the percentage of hypocretin cells expressing c-Fos in the perifornical, dorsomedial, and lateral hypothalamus. Nicotine 0-8 hypocretin Mus musculus 48-58 21831361-11 2012 CONCLUSIONS: These data demonstrate that hypocretin signaling acting on Hcrtr-1 in the PVN plays a crucial role in the expression of nicotine withdrawal. Nicotine 133-141 hypocretin Mus musculus 41-51 22293586-0 2012 Role of orexin in the central regulation of glucose and energy homeostasis. Glucose 44-51 hypocretin Mus musculus 8-14 22293586-3 2012 Intriguingly, central administration of orexin is reported to cause blood glucose-elevating effect or blood glucose-lowering effect in rodents, depending on the experimental conditions. Glucose 74-81 hypocretin Mus musculus 40-46 22293586-3 2012 Intriguingly, central administration of orexin is reported to cause blood glucose-elevating effect or blood glucose-lowering effect in rodents, depending on the experimental conditions. Glucose 108-115 hypocretin Mus musculus 40-46 22293586-5 2012 The fact that orexin exhibits biphasic effects on autonomic nerve activity and lipolysis suggests that orexin dually regulates the glucose appearance. Glucose 131-138 hypocretin Mus musculus 14-20 22293586-5 2012 The fact that orexin exhibits biphasic effects on autonomic nerve activity and lipolysis suggests that orexin dually regulates the glucose appearance. Glucose 131-138 hypocretin Mus musculus 103-109 22293586-6 2012 In fact, orexin neurons are activated not only depending on the demand for glucose but also according to a circadian rhythm in the suprachiasmatic nucleus. Glucose 75-82 hypocretin Mus musculus 9-15 22293586-7 2012 The excited orexin neurons appear to alter the sympathetic or parasympathetic outflow to the periphery, and modulate the glucose production and utilization. Glucose 121-128 hypocretin Mus musculus 12-18 22005675-0 2011 Orexin neurons as conditional glucosensors: paradoxical regulation of sugar sensing by intracellular fuels. Sugars 70-75 hypocretin Mus musculus 0-6 22005675-1 2011 Central orexin/hypocretin neurons promote wakefulness, feeding and reward-seeking, and control blood glucose levels by regulating sympathetic outflow to the periphery. Glucose 101-108 hypocretin Mus musculus 8-14 22005675-2 2011 Glucose itself directly suppresses the electrical activity and cytosolic calcium levels of orexin cells. Glucose 0-7 hypocretin Mus musculus 91-97 22005675-2 2011 Glucose itself directly suppresses the electrical activity and cytosolic calcium levels of orexin cells. Calcium 73-80 hypocretin Mus musculus 91-97 22005675-3 2011 Recent in vitro studies suggested that glucose inhibition of orexin cells may be mechanistically unusual, because it persists under conditions where glucose metabolism is unlikely. Glucose 39-46 hypocretin Mus musculus 61-67 22005675-3 2011 Recent in vitro studies suggested that glucose inhibition of orexin cells may be mechanistically unusual, because it persists under conditions where glucose metabolism is unlikely. Glucose 149-156 hypocretin Mus musculus 61-67 22005675-5 2011 Consistent with their documented insensitivity to glucokinase inhibitors, the glucose responses of orexin cells persisted in the presence of the mitochondrial poison oligomycin or the glial toxin fluoroacetate. Glucose 78-85 hypocretin Mus musculus 99-105 22005675-5 2011 Consistent with their documented insensitivity to glucokinase inhibitors, the glucose responses of orexin cells persisted in the presence of the mitochondrial poison oligomycin or the glial toxin fluoroacetate. Oligomycins 166-176 hypocretin Mus musculus 99-105 22005675-5 2011 Consistent with their documented insensitivity to glucokinase inhibitors, the glucose responses of orexin cells persisted in the presence of the mitochondrial poison oligomycin or the glial toxin fluoroacetate. Fluoroacetates 196-209 hypocretin Mus musculus 99-105 22005675-11 2011 We propose that this unexpected intrinsic property of orexin cells allows them to act as "conditional glucosensors" that preferentially respond to glucose during reduced background energy levels. Glucose 102-109 hypocretin Mus musculus 54-60 21911618-7 2011 Nifedipine (1 muM), as well as 2-aminoethyl diphenyl borate (10 muM), reduced the amplitude and shortened the duration of the response to OXA, which was abolished by Ni(2+) (50 muM) or TEA (1 mM). Nifedipine 0-10 hypocretin Mus musculus 138-141 21911618-7 2011 Nifedipine (1 muM), as well as 2-aminoethyl diphenyl borate (10 muM), reduced the amplitude and shortened the duration of the response to OXA, which was abolished by Ni(2+) (50 muM) or TEA (1 mM). 2-aminoethyldiphenylborate 31-59 hypocretin Mus musculus 138-141 21727218-4 2011 By using "ATP clamp" we demonstrated that membrane potential (V(m)) correlated with the [ATP](i) in Hcrt neurones. Adenosine Triphosphate 10-13 hypocretin Mus musculus 100-104 21727218-4 2011 By using "ATP clamp" we demonstrated that membrane potential (V(m)) correlated with the [ATP](i) in Hcrt neurones. Adenosine Triphosphate 89-92 hypocretin Mus musculus 100-104 21727218-6 2011 A direct disruption of ATP production or reduction in ambient glucose levels resulted in an inhibition of activity in Hcrt neurones. Adenosine Triphosphate 23-26 hypocretin Mus musculus 118-122 21727218-6 2011 A direct disruption of ATP production or reduction in ambient glucose levels resulted in an inhibition of activity in Hcrt neurones. Glucose 62-69 hypocretin Mus musculus 118-122 21727218-7 2011 The V(m) was significantly depolarized in Hcrt neurones in sleep-deprived mice as compared with controls (P < 0.01, t test), which was eliminated by experimental manipulations causing the same level of [ATP](i) and K(ATP) channel opening in both groups, suggesting a decrease during sleep and an increase during sustained wakefulness in [ATP](i) in Hcrt cells. Adenosine Triphosphate 220-223 hypocretin Mus musculus 42-46 21727218-7 2011 The V(m) was significantly depolarized in Hcrt neurones in sleep-deprived mice as compared with controls (P < 0.01, t test), which was eliminated by experimental manipulations causing the same level of [ATP](i) and K(ATP) channel opening in both groups, suggesting a decrease during sleep and an increase during sustained wakefulness in [ATP](i) in Hcrt cells. Adenosine Triphosphate 220-223 hypocretin Mus musculus 42-46 21324360-7 2011 Nevertheless, even at advanced stages of HD, intrinsic firing properties of orexin cells remain normal and suppressible by serotonin, noradrenaline, and glucose. Serotonin 123-132 hypocretin Mus musculus 76-82 21324360-7 2011 Nevertheless, even at advanced stages of HD, intrinsic firing properties of orexin cells remain normal and suppressible by serotonin, noradrenaline, and glucose. Norepinephrine 134-147 hypocretin Mus musculus 76-82 21324360-7 2011 Nevertheless, even at advanced stages of HD, intrinsic firing properties of orexin cells remain normal and suppressible by serotonin, noradrenaline, and glucose. Glucose 153-160 hypocretin Mus musculus 76-82 21415167-7 2011 Most interestingly, orexin-A also promoted robust apoptosis in cells that are resistant to the most commonly used drug in colon cancer chemotherapy, 5-fluorouracil. Fluorouracil 149-163 hypocretin Mus musculus 20-28 21107540-0 2011 Involvement of the orexin/hypocretin system in ethanol conditioned place preference. Ethanol 47-54 hypocretin Mus musculus 19-25 21107540-1 2011 RATIONALE: Recent studies suggest that orexin/hypocretin is involved in drug reward and drug-seeking behaviors, including ethanol self-administration. Ethanol 122-129 hypocretin Mus musculus 39-45 21107540-1 2011 RATIONALE: Recent studies suggest that orexin/hypocretin is involved in drug reward and drug-seeking behaviors, including ethanol self-administration. Ethanol 122-129 hypocretin Mus musculus 46-56 21107540-2 2011 However, orexin"s role in ethanol-induced seeking behaviors remains unclear. Ethanol 26-33 hypocretin Mus musculus 9-15 21107540-3 2011 OBJECTIVE: These studies examined the role of orexin in the acquisition and expression of ethanol conditioned place preference (CPP) using the orexin 1 receptor (OX1R) antagonist SB-334867. Ethanol 90-97 hypocretin Mus musculus 46-52 21464907-6 2011 Using the CR model, we then attempted to verify the published result that orexin deletion diminishes food anticipatory activity (FAA) but observed little to no diminution in the response to CR and, surprisingly, that orexin KO mice are refractory to body weight loss on a CR diet. Chromium 10-12 hypocretin Mus musculus 74-80 21258173-1 2011 Orexin-A is a newly identified neuropeptide expressed in the lateral areas of the hypothalamus that plays a role in various physiological functions, including regulation of glucose metabolism. Glucose 173-180 hypocretin Mus musculus 0-8 21949857-5 2011 The number of Fos-positive orexin neurons markedly decreased after intraperitoneal administration of glycine to mice. Glycine 101-108 hypocretin Mus musculus 27-33 21949857-7 2011 Glycine directly induced hyperpolarization and cessation of firing of orexin neurons. Glycine 0-7 hypocretin Mus musculus 70-76 21086064-0 2011 Decreased intake of sucrose solutions in orexin knockout mice. Sucrose 20-27 hypocretin Mus musculus 41-47 21086064-8 2011 Orexin deficiency may lower the satiety threshold resulting in reduced sucrose intake, without altering food intake. Sucrose 71-78 hypocretin Mus musculus 0-6 21258173-5 2011 Intracerebroventricular administration of orexin-A (2.5, 25, or 250 pmol/mouse) significantly and dose-dependently suppressed the development of post-ischemic glucose intolerance on day 1 after MCAO and neuronal damage on day 3 after MCAO. Glucose 159-166 hypocretin Mus musculus 42-50 21258173-7 2011 Furthermore, these expressions were completely recovered to normal levels by orexin-A and were reversed by the administration of SB334867, a specific orexin-1 receptor antagonist. 1-(2-methylbenzoxazol-6-yl)-3-(1,5)naphthyridin-4-yl urea 129-137 hypocretin Mus musculus 77-85 21258173-8 2011 These results suggest that regulation of post-ischemic glucose intolerance by orexin-A suppressed cerebral ischemic neuronal damage. Glucose 55-62 hypocretin Mus musculus 78-86 20587130-0 2010 Hypothalamic orexin and pro-opiomelanocortin activities are essential for the anorexic effects of m-chlorophenylpiperazine in mice. 1-(3-chlorophenyl)piperazine 98-122 hypocretin Mus musculus 13-19 20696213-3 2010 Modafinil is also thought to express its effect through the orexinergic neurons, and orexin increases hypothalamic histamine release. Modafinil 0-9 hypocretin Mus musculus 60-66 20696213-5 2010 In the present study, we performed in vivo microdialysis and c-Fos immunohistochemistry to investigate whether the orexinergic system mediates the activation of the histaminergic system by modafinil using orexin neuron-deficient mice. Modafinil 189-198 hypocretin Mus musculus 115-121 20587130-5 2010 The injection of orexin siRNA oligonucleotides suppressed the hyperphagia induced by the injection of POMC siRNA oligonucleotides. Oligonucleotides 30-46 hypocretin Mus musculus 17-23 20587130-5 2010 The injection of orexin siRNA oligonucleotides suppressed the hyperphagia induced by the injection of POMC siRNA oligonucleotides. Oligonucleotides 113-129 hypocretin Mus musculus 17-23 20587130-6 2010 These findings suggest that functional hypothalamic POMC and orexin activity has a critical role in satiety signalling of mCPP in mice. 1-(3-chlorophenyl)piperazine 122-126 hypocretin Mus musculus 61-67 20620197-5 2010 Contrary to the expectation, injection of QA (60 and 120 nmol) into the lateral hypothalamus of male C57BL/6 mice caused selective loss of MCH neurons rather than orexin neurons, and this toxicity of QA was attenuated by MK-801, an NMDA receptor antagonist. Quinolinic Acid 42-44 hypocretin Mus musculus 163-169 20587130-2 2010 Here we show that food-restricted wild-type mice, which exhibited decreased hypothalamic POMC expression and increased hypothalamic orexin expression, were responsive to m-chlorophenylpiperazine (m-CPP), a 5-HT(2C/1B) receptor agonist, leading to anorexia, whereas food-restricted A(y) mice with decreased hypothalamic POMC and orexin expression, were not. 1-(3-chlorophenyl)piperazine 170-194 hypocretin Mus musculus 132-138 20587130-2 2010 Here we show that food-restricted wild-type mice, which exhibited decreased hypothalamic POMC expression and increased hypothalamic orexin expression, were responsive to m-chlorophenylpiperazine (m-CPP), a 5-HT(2C/1B) receptor agonist, leading to anorexia, whereas food-restricted A(y) mice with decreased hypothalamic POMC and orexin expression, were not. 1-(3-chlorophenyl)piperazine 170-194 hypocretin Mus musculus 328-334 20587130-2 2010 Here we show that food-restricted wild-type mice, which exhibited decreased hypothalamic POMC expression and increased hypothalamic orexin expression, were responsive to m-chlorophenylpiperazine (m-CPP), a 5-HT(2C/1B) receptor agonist, leading to anorexia, whereas food-restricted A(y) mice with decreased hypothalamic POMC and orexin expression, were not. 1-(3-chlorophenyl)piperazine 196-201 hypocretin Mus musculus 132-138 20587130-2 2010 Here we show that food-restricted wild-type mice, which exhibited decreased hypothalamic POMC expression and increased hypothalamic orexin expression, were responsive to m-chlorophenylpiperazine (m-CPP), a 5-HT(2C/1B) receptor agonist, leading to anorexia, whereas food-restricted A(y) mice with decreased hypothalamic POMC and orexin expression, were not. 1-(3-chlorophenyl)piperazine 196-201 hypocretin Mus musculus 328-334 20587130-3 2010 Injection of POMC small interfering RNA (siRNA) oligonucleotide+orexin siRNA oligonucleotide into the third cerebral ventricle was unresponsive to mCPP-induced anorexia, whereas a single injection of POMC or orexin siRNA oligonucleotides elicited a response. Oligonucleotides 77-92 hypocretin Mus musculus 64-70