PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 15673672-5 2005 MCH1r ko mice exhibited robust hyperactivity in a novel or familiar environment and were super-sensitive to the locomotor activating effects of d-amphetamine and the D1 agonist 2,3,4,5-tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benazepine HCl. Dextroamphetamine 144-157 melanin-concentrating hormone receptor 1 Mus musculus 0-5 15890841-0 2005 cAMP and extracellular signal-regulated kinase signaling in response to d-amphetamine and methylphenidate in the prefrontal cortex in vivo: role of beta 1-adrenoceptors. Dextroamphetamine 72-85 mitogen-activated protein kinase 1 Mus musculus 9-46 15890841-4 2005 To investigate the intracellular signaling pathways activated by d-amphetamine and methylphenidate in the prefrontal cortex in vivo in mice, we measured the cAMP-dependent Ser845 phosphorylation of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor GluR1 subunit and the active form of extracellular signal-regulated kinase (ERK). Dextroamphetamine 65-78 glutamate receptor, ionotropic, AMPA1 (alpha 1) Mus musculus 264-269 15890841-4 2005 To investigate the intracellular signaling pathways activated by d-amphetamine and methylphenidate in the prefrontal cortex in vivo in mice, we measured the cAMP-dependent Ser845 phosphorylation of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor GluR1 subunit and the active form of extracellular signal-regulated kinase (ERK). Dextroamphetamine 65-78 mitogen-activated protein kinase 1 Mus musculus 301-338 15890841-4 2005 To investigate the intracellular signaling pathways activated by d-amphetamine and methylphenidate in the prefrontal cortex in vivo in mice, we measured the cAMP-dependent Ser845 phosphorylation of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor GluR1 subunit and the active form of extracellular signal-regulated kinase (ERK). Dextroamphetamine 65-78 mitogen-activated protein kinase 1 Mus musculus 340-343 15890841-5 2005 Administration of d-amphetamine (5-10 mg/kg) or methylphenidate (10-20 mg/kg) increased phosphorylation of GluR1. Dextroamphetamine 18-31 glutamate receptor, ionotropic, AMPA1 (alpha 1) Mus musculus 107-112 15890841-6 2005 Basal and d-amphetamine-induced GluR1 phosphorylation was reduced by propranolol, a general beta-adrenoceptor antagonist, and betaxolol, a beta1-antagonist, but not by (+/-)-1-[2,3-(dihydro-7-methyl-1H-inden-4-yl)oxy]-3-[(1-methylethyl)amino]-2-butanol (ICI-118,515), a beta2-antagonist. Dextroamphetamine 10-23 glutamate receptor, ionotropic, AMPA1 (alpha 1) Mus musculus 32-37 15890841-6 2005 Basal and d-amphetamine-induced GluR1 phosphorylation was reduced by propranolol, a general beta-adrenoceptor antagonist, and betaxolol, a beta1-antagonist, but not by (+/-)-1-[2,3-(dihydro-7-methyl-1H-inden-4-yl)oxy]-3-[(1-methylethyl)amino]-2-butanol (ICI-118,515), a beta2-antagonist. Dextroamphetamine 10-23 histocompatibility 2, O region beta locus Mus musculus 268-275 15890841-10 2005 d-amphetamine but not methylphenidate increased ERK phosphorylation. Dextroamphetamine 0-13 mitogen-activated protein kinase 1 Mus musculus 48-51 15680173-2 2005 We reported that, compared to Long-Evans (LEH) rats, Sprague-Dawley (SDH) rats are more sensitive to the PPI-disruptive effects of the direct D1/D2 agonist apomorphine (APO) and the indirect DA agonist d-amphetamine (AMPH). Dextroamphetamine 202-215 serine dehydratase Rattus norvegicus 69-72 15680173-2 2005 We reported that, compared to Long-Evans (LEH) rats, Sprague-Dawley (SDH) rats are more sensitive to the PPI-disruptive effects of the direct D1/D2 agonist apomorphine (APO) and the indirect DA agonist d-amphetamine (AMPH). Dextroamphetamine 217-221 serine dehydratase Rattus norvegicus 69-72 15680173-5 2005 SDH rats were more sensitive than LEH rats to the PPI-disruptive effects of both APO (0.5 mg/kg) and AMPH (4.5 mg/kg). Dextroamphetamine 101-105 serine dehydratase Rattus norvegicus 0-3 15608059-4 2005 Here, we show in mouse that d-amphetamine activates ERK in a subset of medium-size spiny neurons of the dorsal striatum and nucleus accumbens, through the combined action of glutamate NMDA and D1-dopamine receptors. Dextroamphetamine 28-41 mitogen-activated protein kinase 1 Mus musculus 52-55 15608059-5 2005 Activation of ERK by d-amphetamine or by widely abused drugs, including cocaine, nicotine, morphine, and Delta(9)-tetrahydrocannabinol was absent in mice lacking dopamine- and cAMP-regulated phosphoprotein of M(r) 32,000 (DARPP-32). Dextroamphetamine 21-34 mitogen-activated protein kinase 1 Mus musculus 14-17 15608059-5 2005 Activation of ERK by d-amphetamine or by widely abused drugs, including cocaine, nicotine, morphine, and Delta(9)-tetrahydrocannabinol was absent in mice lacking dopamine- and cAMP-regulated phosphoprotein of M(r) 32,000 (DARPP-32). Dextroamphetamine 21-34 protein phosphatase 1, regulatory inhibitor subunit 1B Mus musculus 222-230 15608059-6 2005 The effects of d-amphetamine or cocaine on ERK activation in the striatum, but not in the prefrontal cortex, were prevented by point mutation of Thr-34, a DARPP-32 residue specifically involved in protein phosphatase-1 inhibition. Dextroamphetamine 15-28 mitogen-activated protein kinase 1 Mus musculus 43-46 15608059-6 2005 The effects of d-amphetamine or cocaine on ERK activation in the striatum, but not in the prefrontal cortex, were prevented by point mutation of Thr-34, a DARPP-32 residue specifically involved in protein phosphatase-1 inhibition. Dextroamphetamine 15-28 protein phosphatase 1, regulatory inhibitor subunit 1B Mus musculus 155-163 15634764-8 2005 d-Amphetamine enhanced SERT basal phosphorylation and PD169316 blocked this effect. Dextroamphetamine 0-13 solute carrier family 6 member 4 Homo sapiens 23-27 15039766-8 2004 Brain NE stores of alpha2A-KO mice were depleted by D-amph, revealing the alpha2A-AR as essential in modulating the actions of D-amph. Dextroamphetamine 127-133 adrenergic receptor, alpha 2a Mus musculus 74-84 15579162-7 2004 Accordingly, the concomitant blockade of 5-HT2A and alpha1b-adrenergic receptors in WT mice entirely blocked acute locomotor responses but also the development of behavioural sensitization to morphine, D-amphetamine or cocaine (10 mg/kg). Dextroamphetamine 202-215 5-hydroxytryptamine (serotonin) receptor 2A Mus musculus 41-47 15579162-7 2004 Accordingly, the concomitant blockade of 5-HT2A and alpha1b-adrenergic receptors in WT mice entirely blocked acute locomotor responses but also the development of behavioural sensitization to morphine, D-amphetamine or cocaine (10 mg/kg). Dextroamphetamine 202-215 calcium channel, voltage-dependent, N type, alpha 1B subunit Mus musculus 52-59 14726993-13 2004 CONCLUSIONS: Our data suggest that mGluR5 receptors not only mediate spontaneous locomotor activity in DBA/2J mice but also the acute locomotor stimulant effects of cocaine, D-amphetamine and lower doses of GBR12909. Dextroamphetamine 174-187 glutamate receptor, ionotropic, kainate 1 Mus musculus 35-41 14574438-0 2004 D-amphetamine responses in catechol-O-methyltransferase (COMT) disrupted mice. Dextroamphetamine 0-13 catechol-O-methyltransferase Mus musculus 27-55 15152888-0 2004 Dextroamphetamine use during B-2 combat missions. Dextroamphetamine 0-17 immunoglobulin kappa variable 5-2 Homo sapiens 29-32 15152888-15 2004 CONCLUSIONS: B-2 pilots in long-duration combat flight selectively employ dextroamphetamine, naps, and other fatigue countermeasures. Dextroamphetamine 74-91 immunoglobulin kappa variable 5-2 Homo sapiens 13-16 15044042-1 2004 The ability of repeated D-amphetamine (2 mg/kg) treatment to induce behavioral sensitization in rats and alter glial fibrillary acidic protein (GFAP), dopamine transporter (DAT) and glutamate transporter-1 (GLT-1) immunoreactivities was assessed after a 10-day drug abstinence period. Dextroamphetamine 24-37 glial fibrillary acidic protein Rattus norvegicus 111-142 15044042-1 2004 The ability of repeated D-amphetamine (2 mg/kg) treatment to induce behavioral sensitization in rats and alter glial fibrillary acidic protein (GFAP), dopamine transporter (DAT) and glutamate transporter-1 (GLT-1) immunoreactivities was assessed after a 10-day drug abstinence period. Dextroamphetamine 24-37 glial fibrillary acidic protein Rattus norvegicus 144-148 15044042-1 2004 The ability of repeated D-amphetamine (2 mg/kg) treatment to induce behavioral sensitization in rats and alter glial fibrillary acidic protein (GFAP), dopamine transporter (DAT) and glutamate transporter-1 (GLT-1) immunoreactivities was assessed after a 10-day drug abstinence period. Dextroamphetamine 24-37 solute carrier family 6 member 3 Rattus norvegicus 151-171 15044042-1 2004 The ability of repeated D-amphetamine (2 mg/kg) treatment to induce behavioral sensitization in rats and alter glial fibrillary acidic protein (GFAP), dopamine transporter (DAT) and glutamate transporter-1 (GLT-1) immunoreactivities was assessed after a 10-day drug abstinence period. Dextroamphetamine 24-37 solute carrier family 6 member 3 Rattus norvegicus 173-176 15044042-1 2004 The ability of repeated D-amphetamine (2 mg/kg) treatment to induce behavioral sensitization in rats and alter glial fibrillary acidic protein (GFAP), dopamine transporter (DAT) and glutamate transporter-1 (GLT-1) immunoreactivities was assessed after a 10-day drug abstinence period. Dextroamphetamine 24-37 solute carrier family 1 member 2 Rattus norvegicus 182-205 15044042-1 2004 The ability of repeated D-amphetamine (2 mg/kg) treatment to induce behavioral sensitization in rats and alter glial fibrillary acidic protein (GFAP), dopamine transporter (DAT) and glutamate transporter-1 (GLT-1) immunoreactivities was assessed after a 10-day drug abstinence period. Dextroamphetamine 24-37 solute carrier family 1 member 2 Rattus norvegicus 207-212 14574438-0 2004 D-amphetamine responses in catechol-O-methyltransferase (COMT) disrupted mice. Dextroamphetamine 0-13 catechol-O-methyltransferase Mus musculus 57-61 14574438-6 2004 OBJECTIVE: Since DAT inhibitors and amphetamine apparently have different mechanisms of action, we were interested to see how COMT knockout mice would react to d-amphetamine treatment. Dextroamphetamine 160-173 catechol-O-methyltransferase Mus musculus 126-130 14574438-12 2004 D-amphetamine-induced (10 mg/kg) hyperlocomotion was less suppressed in male COMT knockout mice than in their wild-type counterparts. Dextroamphetamine 0-13 catechol-O-methyltransferase Mus musculus 77-81 14574438-16 2004 In dopaminergic neurons, the contribution of intracellular COMT remains secondary in conditions when dopamine is released by d-amphetamine. Dextroamphetamine 125-138 catechol-O-methyltransferase Mus musculus 59-63 12510024-1 2003 Acute administration of D-amphetamine sulphate (AMPH) and (1-[1-phenylcyclohexyl]piperidine hydrochloride) (phencyclidine; PCP) produces a characteristic spatio-temporal distribution of c-Fos protein in the brain. Dextroamphetamine 24-46 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 186-191 12652343-2 2003 The aim of the present study was to examine whether the development of d-amphetamine (AMPH)-induced sensitisation is related to an altered P2Y(1) receptor expression. Dextroamphetamine 71-84 purinergic receptor P2Y1 Rattus norvegicus 139-145 12652343-2 2003 The aim of the present study was to examine whether the development of d-amphetamine (AMPH)-induced sensitisation is related to an altered P2Y(1) receptor expression. Dextroamphetamine 86-90 purinergic receptor P2Y1 Rattus norvegicus 139-145 12652343-7 2003 Quantification of the P2Y(1) receptor stained cells revealed an increase in the receptor expression after AMPH-induced sensitisation in the studied regions. Dextroamphetamine 106-110 purinergic receptor P2Y1 Rattus norvegicus 22-28 12652343-10 2003 Confocal laser scanning microscopy indicated the localisation of P2Y(1) receptors on GFAP-labelled astrocytes as well as on tubulin (betaIII)-labelled neurones, under control conditions and after AMPH administration. Dextroamphetamine 196-200 purinergic receptor P2Y1 Rattus norvegicus 65-71 12652343-11 2003 CONCLUSION: The present results confirm the existence of P2Y(1) receptors on astrocytes and neurones as possible targets of endogenous ATP and in addition show their up-regulation as a consequence of P2Y(1) receptor-involvement in AMPH-induced sensitisation in vivo. Dextroamphetamine 231-235 purinergic receptor P2Y1 Rattus norvegicus 57-63 12652343-11 2003 CONCLUSION: The present results confirm the existence of P2Y(1) receptors on astrocytes and neurones as possible targets of endogenous ATP and in addition show their up-regulation as a consequence of P2Y(1) receptor-involvement in AMPH-induced sensitisation in vivo. Dextroamphetamine 231-235 purinergic receptor P2Y1 Rattus norvegicus 200-206 12631566-7 2003 However, when cis-flupenthixol and D-amphetamine were present in the mPFC during presentation of the tone alone, freezing to the tone was reduced. Dextroamphetamine 35-48 complement factor properdin Mus musculus 69-73 12631566-10 2003 The reduction of conditioned fear by prefrontal infusion of both cis-flupenthixol and D-amphetamine may reflect normal expression of conditioned fear requires an optimal level of mPFC dopamine activity. Dextroamphetamine 86-99 complement factor properdin Mus musculus 179-183 14556232-1 2003 Recent experiments have shown that mice lacking the alpha1b-adrenergic receptor (alpha1b-AR KO) are less responsive to the locomotor hyperactivity induced by psychostimulants, such as D-amphetamine or cocaine, than their wild-type littermates (WT). Dextroamphetamine 184-197 adrenergic receptor, alpha 1b Mus musculus 52-79 14556232-1 2003 Recent experiments have shown that mice lacking the alpha1b-adrenergic receptor (alpha1b-AR KO) are less responsive to the locomotor hyperactivity induced by psychostimulants, such as D-amphetamine or cocaine, than their wild-type littermates (WT). Dextroamphetamine 184-197 calcium channel, voltage-dependent, N type, alpha 1B subunit Mus musculus 52-59 14599485-3 2003 A significant restoration (P<0.01) in D-amphetamine induced rotations, spontaneous and apomorphine induced locomotor activity in rats co-transplanted with VMC and GDNF was observed as compared to VMC alone transplanted rats. Dextroamphetamine 41-54 glial cell derived neurotrophic factor Rattus norvegicus 166-170 12801594-4 2003 The neuropeptides substance P (SP) and cholecystokinin (CCK) are present in the striatum where they could play an important role regulating the effects of psychostimulants like cocaine and amphetamines (methamphetamine [METH] is a long acting derivative of d-amphetamine). Dextroamphetamine 257-270 cholecystokinin Rattus norvegicus 39-54 12801594-4 2003 The neuropeptides substance P (SP) and cholecystokinin (CCK) are present in the striatum where they could play an important role regulating the effects of psychostimulants like cocaine and amphetamines (methamphetamine [METH] is a long acting derivative of d-amphetamine). Dextroamphetamine 257-270 cholecystokinin Rattus norvegicus 56-59 12510024-1 2003 Acute administration of D-amphetamine sulphate (AMPH) and (1-[1-phenylcyclohexyl]piperidine hydrochloride) (phencyclidine; PCP) produces a characteristic spatio-temporal distribution of c-Fos protein in the brain. Dextroamphetamine 48-52 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 186-191 12510024-8 2003 Finally, supershift analysis clearly demonstrated presence of SRF and c-Fos protein in the transcriptional complexes bound to SRE and DSE sequences irrespective to AMPH, PCP or combined treatment. Dextroamphetamine 164-168 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 70-75 12596507-0 2002 [Locomotor activity and expression of c-Fos protein in the brain of C57BL and Balb/c mice: effects of D-amphetamine and sydnocarb]. Dextroamphetamine 102-115 FBJ osteosarcoma oncogene Mus musculus 38-43 12423250-1 2002 In heterologous expression systems, dopamine transporter (DAT) cell-surface localization is reduced after relatively prolonged exposure to d-amphetamine (AMPH) or dopamine (DA), suggesting a role for substrate-mediated regulation of transporter function. Dextroamphetamine 139-152 solute carrier family 6 member 3 Rattus norvegicus 36-56 12423250-1 2002 In heterologous expression systems, dopamine transporter (DAT) cell-surface localization is reduced after relatively prolonged exposure to d-amphetamine (AMPH) or dopamine (DA), suggesting a role for substrate-mediated regulation of transporter function. Dextroamphetamine 139-152 solute carrier family 6 member 3 Rattus norvegicus 58-61 12423250-1 2002 In heterologous expression systems, dopamine transporter (DAT) cell-surface localization is reduced after relatively prolonged exposure to d-amphetamine (AMPH) or dopamine (DA), suggesting a role for substrate-mediated regulation of transporter function. Dextroamphetamine 154-158 solute carrier family 6 member 3 Rattus norvegicus 36-56 12423250-1 2002 In heterologous expression systems, dopamine transporter (DAT) cell-surface localization is reduced after relatively prolonged exposure to d-amphetamine (AMPH) or dopamine (DA), suggesting a role for substrate-mediated regulation of transporter function. Dextroamphetamine 154-158 solute carrier family 6 member 3 Rattus norvegicus 58-61 12423250-3 2002 In human DAT-expressing Xenopus laevis oocytes, repeated exposure to low micromolar concentrations of DA, AMPH or tyramine markedly reduced transport-mediated currents. Dextroamphetamine 106-110 solute carrier family 6 member 3 Homo sapiens 9-12 12498914-2 2003 Previously we showed that one of our brain-penetrating neurotensin analogs, NT69L (N-met-L-Arg, L-Lys, L-Pro, L-neo-Trp, L-tert-Leu, L-Leu), blocks cocaine- and D-amphetamine-induced hyperactivity in rats. Dextroamphetamine 161-174 neurotensin Rattus norvegicus 55-66 15035814-0 2003 Interactions of D-amphetamine with the active site of monoamine oxidase-A. Dextroamphetamine 16-29 monoamine oxidase A Homo sapiens 54-73 12445480-5 2002 The inhibitors, D-amphetamine, harmine, tetrindole, and befloxatone all induce similar (but not identical) changes in the spectrum of MAO A, consistent with stacking of inhibitor with the flavin in the active site. Dextroamphetamine 16-29 monoamine oxidase A Homo sapiens 134-139 12445480-6 2002 D-Amphetamine, harmine, and tetrindole stabilise the semiquinone form of FAD during reduction of MAO A by dithionite and no further reduction of these inhibitor-MAO A complexes has been observed. Dextroamphetamine 0-13 monoamine oxidase A Homo sapiens 97-102 12393054-3 2002 In the present study, we examined the effects of the indirect psychostimulant, D-amphetamine, on (CRP40) expression in discrete brain regions. Dextroamphetamine 79-92 heat shock protein family A (Hsp70) member 9 Rattus norvegicus 98-103 12393054-4 2002 The technique of Western immunoblotting was utilized for quantitation of CRP40 in different experimental paradigms following D-amphetamine treatment. Dextroamphetamine 125-138 heat shock protein family A (Hsp70) member 9 Rattus norvegicus 73-78 12393054-12 2002 The results of this study demonstrate selective modulation of CRP40 by D-amphetamine treatment, without affecting the 70-kDa heat shock protein. Dextroamphetamine 71-84 heat shock protein family A (Hsp70) member 9 Rattus norvegicus 62-67 12596507-1 2002 Effects of the psychostimulants D-amphetamine and sydnocarb on the locomotor activity and c-Fos protein expression in the brain of C57BL and BALB/c mice was studied. Dextroamphetamine 32-45 FBJ osteosarcoma oncogene Mus musculus 90-95 12596507-3 2002 The effect of D-amphetamine with respect to the locomotor activity was more pronounced in C57BL mice, which correlates with a higher level of the c-Fos expression in the secondary motor cortex. Dextroamphetamine 14-27 FBJ osteosarcoma oncogene Mus musculus 146-151 12596507-4 2002 At the same time syndocarb more significantly than D-amphetamine induces expression of the c-Fos protein in the nucleus accumbens core of C57BL mice. Dextroamphetamine 51-64 FBJ osteosarcoma oncogene Mus musculus 91-96 12111825-0 2002 d-Amphetamine-induced increase in neurotensin and neuropeptide Y outflow in the ventral striatum is mediated via stimulation of dopamine D1 and D2/3 receptors. Dextroamphetamine 0-13 neurotensin Rattus norvegicus 34-45 12111825-0 2002 d-Amphetamine-induced increase in neurotensin and neuropeptide Y outflow in the ventral striatum is mediated via stimulation of dopamine D1 and D2/3 receptors. Dextroamphetamine 0-13 neuropeptide Y Rattus norvegicus 50-64 12111825-4 2002 d-Amphetamine significantly increased extracellular NT- and NPY-like immunoreactivity (LI) concentrations compared with control animals. Dextroamphetamine 0-13 neuropeptide Y Rattus norvegicus 60-63 12111825-7 2002 These findings demonstrate that d-amphetamine increases extracellular concentrations of NT-LI and NPY-LI in the VSTR through a mechanism that initially involves stimulation of either DA-D(1) or DA-D(2/3) receptors but appears to require both. Dextroamphetamine 32-45 neuropeptide Y Rattus norvegicus 98-101 11525773-0 2001 A novel neurotensin analog blocks cocaine- and D-amphetamine-induced hyperactivity. Dextroamphetamine 47-60 neurotensin Homo sapiens 8-19 12105094-5 2002 The expression of c-fos protein in striatal neurons was much more increased after a single injection of D-AMPH (5 mg/kg) than after an equimolar concentration of SYD (23.8 mg/kg) in both the anterior and the posterior part of the striatum. Dextroamphetamine 104-110 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 18-23 11797063-3 2001 OBJECTIVES: The goal of the present study was to investigate the effects of acute and daily administration of d-amphetamine on the locomotor activity of DAT(-/-) mice and examine the development of behavioral sensitization. Dextroamphetamine 110-123 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 Mus musculus 153-156 11797063-7 2001 RESULTS: Acute and repeated d-amphetamine injection (1 mg/kg) induced an hypolocomotor response in DAT(-/-) and DAT(+/-) mice, but only DAT(+/-) mice developed locomotor sensitization to the drug. Dextroamphetamine 28-41 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 Mus musculus 99-102 11797063-7 2001 RESULTS: Acute and repeated d-amphetamine injection (1 mg/kg) induced an hypolocomotor response in DAT(-/-) and DAT(+/-) mice, but only DAT(+/-) mice developed locomotor sensitization to the drug. Dextroamphetamine 28-41 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 Mus musculus 112-115 11797063-7 2001 RESULTS: Acute and repeated d-amphetamine injection (1 mg/kg) induced an hypolocomotor response in DAT(-/-) and DAT(+/-) mice, but only DAT(+/-) mice developed locomotor sensitization to the drug. Dextroamphetamine 28-41 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 Mus musculus 112-115 11797063-9 2001 The common hypolocomotor effect induced by d-amphetamine and fluoxetine in DAT(-/-) mice suggests an action on the serotonin transporter. Dextroamphetamine 43-56 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 Mus musculus 75-78 11797063-11 2001 CONCLUSIONS: These findings indicate that partial or total DAT gene deletion result in decreased locomotion in response to d-amphetamine and modify behavioral sensitization depending on the proportion of DAT removed, suggesting that inhibition of the DAT is necessary for the development of sensitization to psychostimulant drugs. Dextroamphetamine 123-136 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 Mus musculus 59-62 11923452-4 2002 Here we show that the locomotor hyperactivities induced by d-amphetamine (1-3 mg/kg), cocaine (5-20 mg/kg), or morphine (5-10 mg/kg) in mice lacking the alpha1b subtype of adrenergic receptors were dramatically decreased when compared with wild-type littermates. Dextroamphetamine 59-72 calcium channel, voltage-dependent, N type, alpha 1B subunit Mus musculus 153-160 11850143-9 2002 The augmented release of CORT and ACTH observed in d-amphetamine-treated rats might have important implications for human disorders in which processes resembling neurochemical sensitization have been hypothesized to play an etiological role. Dextroamphetamine 51-64 cortistatin Rattus norvegicus 25-29 11900599-3 2001 In this study, the activities of several antioxidant enzymes in different areas of rat brain were measured after repeated administration of d-amphetamine sulphate (sc, 20 mg/kg/day, for 14 days), namely glutathione-S-transferase (GST), glutathione peroxidase (GPx), glutathione reductase (GRed), catalase, and superoxide dismutase (SOD). Dextroamphetamine 140-162 hematopoietic prostaglandin D synthase Rattus norvegicus 230-233 11900599-3 2001 In this study, the activities of several antioxidant enzymes in different areas of rat brain were measured after repeated administration of d-amphetamine sulphate (sc, 20 mg/kg/day, for 14 days), namely glutathione-S-transferase (GST), glutathione peroxidase (GPx), glutathione reductase (GRed), catalase, and superoxide dismutase (SOD). Dextroamphetamine 140-162 glutathione-disulfide reductase Rattus norvegicus 266-287 11900599-3 2001 In this study, the activities of several antioxidant enzymes in different areas of rat brain were measured after repeated administration of d-amphetamine sulphate (sc, 20 mg/kg/day, for 14 days), namely glutathione-S-transferase (GST), glutathione peroxidase (GPx), glutathione reductase (GRed), catalase, and superoxide dismutase (SOD). Dextroamphetamine 140-162 glutathione-disulfide reductase Rattus norvegicus 289-293 11900599-3 2001 In this study, the activities of several antioxidant enzymes in different areas of rat brain were measured after repeated administration of d-amphetamine sulphate (sc, 20 mg/kg/day, for 14 days), namely glutathione-S-transferase (GST), glutathione peroxidase (GPx), glutathione reductase (GRed), catalase, and superoxide dismutase (SOD). Dextroamphetamine 140-162 catalase Rattus norvegicus 296-304 11900599-4 2001 When compared to a pair-fed control group, d-amphetamine treatment enhanced the activity of GST in hypothalamus to 167%, GPx in striatum to 127%, in nucleus accumbens to 192%, and in medial prefrontal cortex to 139%, GRed in hypothalamus to 139%, as well as catalase in medial prefrontal cortex to 153%. Dextroamphetamine 43-56 hematopoietic prostaglandin D synthase Rattus norvegicus 92-95 11900599-4 2001 When compared to a pair-fed control group, d-amphetamine treatment enhanced the activity of GST in hypothalamus to 167%, GPx in striatum to 127%, in nucleus accumbens to 192%, and in medial prefrontal cortex to 139%, GRed in hypothalamus to 139%, as well as catalase in medial prefrontal cortex to 153%. Dextroamphetamine 43-56 glutathione-disulfide reductase Rattus norvegicus 217-221 11900599-4 2001 When compared to a pair-fed control group, d-amphetamine treatment enhanced the activity of GST in hypothalamus to 167%, GPx in striatum to 127%, in nucleus accumbens to 192%, and in medial prefrontal cortex to 139%, GRed in hypothalamus to 139%, as well as catalase in medial prefrontal cortex to 153%. Dextroamphetamine 43-56 catalase Rattus norvegicus 258-266 11398184-0 2001 Dopamine receptor antagonists prevent the d-amphetamine-induced increase in calcitonin gene-related peptide levels in ventral striatum. Dextroamphetamine 42-55 calcitonin-related polypeptide alpha Rattus norvegicus 76-107 11398184-1 2001 Microdialysis in conjunction with radioimmunoassay (RIA) were used to study the effects of acute d-amphetamine or dopamine (DA) receptor antagonists administration on extracellular concentrations of calcitonin gene-related peptide (CGRP) in the ventral striatum of the rat. Dextroamphetamine 97-110 calcitonin-related polypeptide alpha Rattus norvegicus 199-230 11398184-4 2001 d-Amphetamine significantly increased extracellular CGRP-LI concentrations compared to the control animals. Dextroamphetamine 0-13 calcitonin-related polypeptide alpha Rattus norvegicus 52-56 11398184-7 2001 The results show that d-amphetamine administration results in an increase in extracellular concentrations of CGRP in the ventral striatum through a mechanism that appears to involve stimulation of either DA-D(1) or DA-D(2/3) receptors. Dextroamphetamine 22-35 calcitonin-related polypeptide alpha Rattus norvegicus 109-113 11440814-6 2001 Despite the administration of haloperidol and D-amphetamine to elicit and enhance neurotensin/neuromedin N messenger RNA expression in striatum, including the nucleus accumbens and olfactory tubercle, no double-labeled neurons were observed there. Dextroamphetamine 46-59 neurotensin Rattus norvegicus 82-106 11163634-6 2001 The specificity of such interaction is supported by the finding that contrary to morphine, cocaine, d-amphetamine and nicotine were self-administered by mice at the same extent either in presence or in absence of the CB1 receptor. Dextroamphetamine 100-113 cannabinoid receptor 1 (brain) Mus musculus 217-220 11329494-1 2001 BACKGROUND: In some individuals, ethanol (EtOH) produces marked stimulant-like subjective effects resembling those of stimulant drugs, like d-amphetamine (AMP). Dextroamphetamine 155-158 adenine phosphoribosyltransferase Homo sapiens 140-153 11337538-3 2001 In this study, we report the effects of oral D-amphetamine relative to placebo on regional cerebral blood flow (rCBF) measured by SPECT in healthy volunteers to characterize the normal CNS response to this primarily dopaminergic stimulant. Dextroamphetamine 45-58 CCAAT/enhancer binding protein zeta Rattus norvegicus 112-116 10889551-2 2000 This neutron-irradiation induced mouse strain is hemizygous for the deletion of the SNAP-25 gene and displays spontaneous hyperactivity that is responsive to dextroamphetamine. Dextroamphetamine 158-175 synaptosomal-associated protein 25 Mus musculus 84-91 10931533-2 2000 An additional aim was to explore the effect of d-amphetamine on NPY-LI release following pretreatment with typical and atypical antipsychotics. Dextroamphetamine 47-60 neuropeptide Y Rattus norvegicus 64-67 10931533-10 2000 d-amphetamine (1.5 mg/kg) significantly increased extracellular NPY-LI in the vehicle group. Dextroamphetamine 0-13 neuropeptide Y Rattus norvegicus 64-67 10931533-12 2000 The results show that d-amphetamine as well as haloperidol and risperidone selectively and specifically affect NPY-LI concentrations in brain tissue and microdialysates and that the effect of d-amphetamine is abolished by both typical and atypical antipsychotics. Dextroamphetamine 22-35 neuropeptide Y Rattus norvegicus 111-114 10931533-12 2000 The results show that d-amphetamine as well as haloperidol and risperidone selectively and specifically affect NPY-LI concentrations in brain tissue and microdialysates and that the effect of d-amphetamine is abolished by both typical and atypical antipsychotics. Dextroamphetamine 192-205 neuropeptide Y Rattus norvegicus 111-114 10578101-3 2000 Dopaminergic stimulation with d-amphetamine or apomorphine induced Fos expression in the central region of the SNC. Dextroamphetamine 30-43 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 67-70 11034152-6 2000 Bursting firing of action potentials in the giant African central RP4 neuron were also elicited after d-amphetamine or l-amphetamine (0.27 mM) administration. Dextroamphetamine 102-115 RGD1559532 Rattus norvegicus 66-69 11034152-15 2000 d-Amphetamine also decreased the calcium, Ia and the steady-state outward currents of the RP4 neuron. Dextroamphetamine 0-13 RGD1559532 Rattus norvegicus 90-93 10733553-5 2000 Four hours after AMPH injection, increase in c-myc and decreases in Bcl-2 and Bcl-X(L) mRNAs occurred in all tissues, whereas p53, Bax, and Bcl-X(S) mRNAs increased in N30 and HCC. Dextroamphetamine 17-21 MYC proto-oncogene, bHLH transcription factor Rattus norvegicus 45-50 10733553-5 2000 Four hours after AMPH injection, increase in c-myc and decreases in Bcl-2 and Bcl-X(L) mRNAs occurred in all tissues, whereas p53, Bax, and Bcl-X(S) mRNAs increased in N30 and HCC. Dextroamphetamine 17-21 BCL2, apoptosis regulator Rattus norvegicus 68-73 10733553-5 2000 Four hours after AMPH injection, increase in c-myc and decreases in Bcl-2 and Bcl-X(L) mRNAs occurred in all tissues, whereas p53, Bax, and Bcl-X(S) mRNAs increased in N30 and HCC. Dextroamphetamine 17-21 Bcl2-like 1 Rattus norvegicus 78-86 10733553-5 2000 Four hours after AMPH injection, increase in c-myc and decreases in Bcl-2 and Bcl-X(L) mRNAs occurred in all tissues, whereas p53, Bax, and Bcl-X(S) mRNAs increased in N30 and HCC. Dextroamphetamine 17-21 Bcl2-like 1 Rattus norvegicus 78-83 10733553-7 2000 c-Myc, P53, Bcl-2, and Bax proteins in normal liver and HCC +/- AMPH showed similar patterns. Dextroamphetamine 64-68 BCL2 associated X, apoptosis regulator Rattus norvegicus 23-26 10733553-9 2000 Basal Hsp27 expression was high in nodules and HCC, and was stimulated by AMPH in liver and N12, but not in N30 and HCC. Dextroamphetamine 74-78 heat shock protein family B (small) member 1 Rattus norvegicus 6-11 10733553-10 2000 These data suggest a role of dysregulation of Bcl-2 family genes and, at least in atypical lesions, of p53 overexpression, in basal and AMPH-induced apoptosis in nodules and HCCs. Dextroamphetamine 136-140 BCL2, apoptosis regulator Rattus norvegicus 46-51 10733553-10 2000 These data suggest a role of dysregulation of Bcl-2 family genes and, at least in atypical lesions, of p53 overexpression, in basal and AMPH-induced apoptosis in nodules and HCCs. Dextroamphetamine 136-140 Wistar clone pR53P1 p53 pseudogene Rattus norvegicus 103-106 10733553-10 2000 These data suggest a role of dysregulation of Bcl-2 family genes and, at least in atypical lesions, of p53 overexpression, in basal and AMPH-induced apoptosis in nodules and HCCs. Dextroamphetamine 136-140 holocytochrome c synthase Rattus norvegicus 174-178 10578101-4 2000 However, lesions of the nigrostriatal dopamine pathway significantly reduced d-amphetamine- and apomorphine-induced Fos expression in the ipsilateral and contralateral SNC, respectively. Dextroamphetamine 77-90 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 116-119 10102769-11 1999 The effect of CP93,129, the most selective of the 5-HT1B agonists, to inhibit the response-potentiating effect of d-amphetamine was reversed by the 5-HT(1B/1D) antagonist GR127935 (3 mg/kg). Dextroamphetamine 114-127 5-hydroxytryptamine receptor 1B Rattus norvegicus 50-56 10486183-2 1999 d-Amphetamine-induced c-fos activation is reduced in the neostriatum deprived of DA afferents. Dextroamphetamine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 10486183-3 1999 Dopaminergic grafts implanted into the denervated neostriatum induce a c-fos hyperexpression when challenged with d-amphetamine, which is correlated with the exaggerated compensation of d-amphetamine-induced rotation. Dextroamphetamine 114-127 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 10486183-3 1999 Dopaminergic grafts implanted into the denervated neostriatum induce a c-fos hyperexpression when challenged with d-amphetamine, which is correlated with the exaggerated compensation of d-amphetamine-induced rotation. Dextroamphetamine 186-199 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 71-76 10486183-7 1999 A c-fos hyperexpression was observed within the grafted neostriatum, which was correlated with the compensation of d-amphetamine- or cocaine-induced rotation. Dextroamphetamine 115-128 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 2-7 10350188-8 1999 d-Amphetamine treatment induced an increase of kidney GST, GRed and catalase levels, and an elevation of N-acetyl-beta-D-glucosaminidase efflux to the urine, accompanied by a decrease in urinary creatinine, compared to the pair-fed group. Dextroamphetamine 0-13 catalase Rattus norvegicus 68-76 9889359-8 1999 In addition, d-amphetamine-evoked overflow of DA was significantly increased in the substantia nigra but not the putamen of MPTP-lesioned monkeys that had received GDNF. Dextroamphetamine 13-26 glial cell derived neurotrophic factor Macaca mulatta 164-168 9833017-0 1997 Effect of CYP2D1 inhibition on the behavioural effects of d-amphetamine. Dextroamphetamine 58-71 cytochrome P450, family 2, subfamily d, polypeptide 1 Rattus norvegicus 10-16 10069519-4 1999 d-Amphetamine-induced Fos expression in rat nucleus accumbens was inhibited by PTAC, thus directly demonstrating the ability of PTAC to modulate DA activity. Dextroamphetamine 0-13 FBJ osteosarcoma oncogene Mus musculus 22-25 9692731-0 1998 D-amphetamine and L-5-hydroxytryptophan-induced behaviours in mice with genetically-altered expression of the alpha2C-adrenergic receptor subtype. Dextroamphetamine 0-13 adrenergic receptor, alpha 2c Mus musculus 110-137 9692731-8 1998 The effect of alpha2C-adrenoceptor gene inactivation was more prominent in D-amphetamine-treated males than in females. Dextroamphetamine 75-88 adrenergic receptor, alpha 2c Mus musculus 14-34 9774240-3 1998 PTAC inhibited conditioned avoidance responding, dopamine receptor agonist-induced behavior and D-amphetamine-induced FOS protein M5 expression in the nucleus accumbens without inducing catalepsy, tremor or salivation at pharmacologically relevant doses. Dextroamphetamine 96-109 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 118-121 9648873-8 1998 Interestingly, d-amphetamine inhibited [3H]DTBZ binding to vesicle membranes with an IC50 of 39.4 microM, a concentration 20 times greater than reported for inhibition of VMAT2 function, suggesting that d-amphetamine interacts with a different site than lobeline on VMAT2 to inhibit monoamine uptake. Dextroamphetamine 15-28 solute carrier family 18 member A2 Rattus norvegicus 171-176 9648873-8 1998 Interestingly, d-amphetamine inhibited [3H]DTBZ binding to vesicle membranes with an IC50 of 39.4 microM, a concentration 20 times greater than reported for inhibition of VMAT2 function, suggesting that d-amphetamine interacts with a different site than lobeline on VMAT2 to inhibit monoamine uptake. Dextroamphetamine 15-28 solute carrier family 18 member A2 Rattus norvegicus 266-271 9648873-8 1998 Interestingly, d-amphetamine inhibited [3H]DTBZ binding to vesicle membranes with an IC50 of 39.4 microM, a concentration 20 times greater than reported for inhibition of VMAT2 function, suggesting that d-amphetamine interacts with a different site than lobeline on VMAT2 to inhibit monoamine uptake. Dextroamphetamine 203-216 solute carrier family 18 member A2 Rattus norvegicus 266-271 10350188-5 1999 d-Amphetamine treatment induced an increase of liver GSH, as well as a decrease of cysteine and MnSOD levels in this organ. Dextroamphetamine 0-13 superoxide dismutase 2 Rattus norvegicus 96-101 10350188-8 1999 d-Amphetamine treatment induced an increase of kidney GST, GRed and catalase levels, and an elevation of N-acetyl-beta-D-glucosaminidase efflux to the urine, accompanied by a decrease in urinary creatinine, compared to the pair-fed group. Dextroamphetamine 0-13 glutathione-disulfide reductase Rattus norvegicus 59-63 20575775-5 1999 Instead, it is argued that inhibition of debrisoquine hydroxylase (cytochrome p450 2D1), an enzyme involved in the metabolism of d-amphetamine, represents the critical mechanism of action. Dextroamphetamine 129-142 cytochrome P450, family 2, subfamily d, polypeptide 1 Rattus norvegicus 67-86 10721048-5 1999 This potentiation was prevented by m-nitrobenzoylalanine, a kynurenine 3-hydroxylase inhibitor that also antagonized the KYNA reduction caused by d-Amph. Dextroamphetamine 146-152 kynurenine 3-monooxygenase Rattus norvegicus 60-84 9786410-0 1998 Distinct and interactive effects of d-amphetamine and haloperidol on levels of neurotensin and its mRNA in subterritories in the dorsal and ventral striatum of the rat. Dextroamphetamine 36-49 neurotensin Rattus norvegicus 79-90 9786410-1 1998 Striatal tissue concentrations of neurotensin, expression of neurotensin/neuromedin N (NT/N) mRNA, and numbers of neurotensin-immunoreactive neurons are increased by d-amphetamine (amph), which stimulates dopamine release in the striatum, and haloperidol (hal), a dopamine receptor antagonist with high affinity for D2-like receptors. Dextroamphetamine 166-179 neurotensin Rattus norvegicus 34-45 9786410-1 1998 Striatal tissue concentrations of neurotensin, expression of neurotensin/neuromedin N (NT/N) mRNA, and numbers of neurotensin-immunoreactive neurons are increased by d-amphetamine (amph), which stimulates dopamine release in the striatum, and haloperidol (hal), a dopamine receptor antagonist with high affinity for D2-like receptors. Dextroamphetamine 166-179 neurotensin Rattus norvegicus 61-85 9786410-1 1998 Striatal tissue concentrations of neurotensin, expression of neurotensin/neuromedin N (NT/N) mRNA, and numbers of neurotensin-immunoreactive neurons are increased by d-amphetamine (amph), which stimulates dopamine release in the striatum, and haloperidol (hal), a dopamine receptor antagonist with high affinity for D2-like receptors. Dextroamphetamine 166-179 neurotensin Rattus norvegicus 87-91 9786410-1 1998 Striatal tissue concentrations of neurotensin, expression of neurotensin/neuromedin N (NT/N) mRNA, and numbers of neurotensin-immunoreactive neurons are increased by d-amphetamine (amph), which stimulates dopamine release in the striatum, and haloperidol (hal), a dopamine receptor antagonist with high affinity for D2-like receptors. Dextroamphetamine 166-179 neurotensin Rattus norvegicus 61-72 9786410-1 1998 Striatal tissue concentrations of neurotensin, expression of neurotensin/neuromedin N (NT/N) mRNA, and numbers of neurotensin-immunoreactive neurons are increased by d-amphetamine (amph), which stimulates dopamine release in the striatum, and haloperidol (hal), a dopamine receptor antagonist with high affinity for D2-like receptors. Dextroamphetamine 168-172 neurotensin Rattus norvegicus 34-45 9786410-1 1998 Striatal tissue concentrations of neurotensin, expression of neurotensin/neuromedin N (NT/N) mRNA, and numbers of neurotensin-immunoreactive neurons are increased by d-amphetamine (amph), which stimulates dopamine release in the striatum, and haloperidol (hal), a dopamine receptor antagonist with high affinity for D2-like receptors. Dextroamphetamine 168-172 neurotensin Rattus norvegicus 61-85 9786410-1 1998 Striatal tissue concentrations of neurotensin, expression of neurotensin/neuromedin N (NT/N) mRNA, and numbers of neurotensin-immunoreactive neurons are increased by d-amphetamine (amph), which stimulates dopamine release in the striatum, and haloperidol (hal), a dopamine receptor antagonist with high affinity for D2-like receptors. Dextroamphetamine 168-172 neurotensin Rattus norvegicus 87-91 9786410-1 1998 Striatal tissue concentrations of neurotensin, expression of neurotensin/neuromedin N (NT/N) mRNA, and numbers of neurotensin-immunoreactive neurons are increased by d-amphetamine (amph), which stimulates dopamine release in the striatum, and haloperidol (hal), a dopamine receptor antagonist with high affinity for D2-like receptors. Dextroamphetamine 168-172 neurotensin Rattus norvegicus 61-72 9804653-8 1998 RESULTS: Both GAP-43 and synaptophysin proteins demonstrated statistically significant increases in density and distribution of immunoreaction product as determined by optical density measurements in the neocortex of the infarcted group treated with D-amphetamines compared with vehicle-treated infarcted controls. Dextroamphetamine 250-264 growth associated protein 43 Rattus norvegicus 14-20 9804653-8 1998 RESULTS: Both GAP-43 and synaptophysin proteins demonstrated statistically significant increases in density and distribution of immunoreaction product as determined by optical density measurements in the neocortex of the infarcted group treated with D-amphetamines compared with vehicle-treated infarcted controls. Dextroamphetamine 250-264 synaptophysin Rattus norvegicus 25-38 9489729-2 1998 Systemic administration of d-amphetamine (2 or 10 mg/kg) increased the striatal output of ACh when the AChE inhibitor neostigmine (0.1 microM) was present in the perfusion fluid. Dextroamphetamine 27-40 acetylcholinesterase (Cartwright blood group) Homo sapiens 103-107 9396872-8 1998 Binding of [99mTc]TRODAT-1 to DAT was found to be inhibited by CFT, methylphenidate, and d-amphetamine in a dose-dependent manner. Dextroamphetamine 89-102 solute carrier family 6 member 3 Rattus norvegicus 21-24 9303570-2 1997 In a previous study, it was shown that a moderate dose of d-amphetamine produced a pattern of EEG power spectrum which indicated a selective activation of D1-like dopamine receptors, whereas a larger dose induced a selective increase of power in the alpha-1 frequency band and, to a lesser degree, in the alpha-2 band, suggesting an additional activation of D2-like receptors. Dextroamphetamine 58-71 adrenoceptor alpha 1D Homo sapiens 250-257 9262339-4 1997 In addition, both potassium- (100 mM, K+) and d-amphetamine (250 microM)-induced DA overflow were augmented in the striatum and nucleus accumbens of the aged rats injected with GDNF. Dextroamphetamine 46-59 glial cell derived neurotrophic factor Rattus norvegicus 177-181 9027869-2 1997 In an attempt to investigate the neurobiological correlates of this phenomenon, the present study examined the effects of prior D-amphetamine sensitization on regional c-fos expression induced by a psychological stressor. Dextroamphetamine 128-141 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-173 9027410-4 1997 Six hours after functional challenge by injection of D-amphetamine (8 mg/kg s.c.), prenatally vehicle-exposed rats exhibited increased preprodynorphin mRNA (ROD) levels in caudate putamen (dorsolateral 187%, dorsomedial 150%, ventrolateral 153%, ventromedial 140% of control), nucleus accumbens (142%) and olfactory tubercle (213%). Dextroamphetamine 53-66 prodynorphin Rattus norvegicus 135-150 8973993-12 1996 Serum prolactin levels were inversely associated with the amount of dextroamphetamine administered, with the largest decrease in serum prolactin levels observed after the 5-mg dose, and this finding was statistically significant. Dextroamphetamine 68-85 prolactin Homo sapiens 6-15 9017269-1 1996 Pretreatment with R(+)-8-OH-DPAT, a selective serotonin (5-HT)1A receptor agonist (50 micrograms/kg, s.c.), inhibited D-amphetamine sulfate (1.0 mg/kg, s.c.)-induced increases in extracellular levels of both 5-HT and dopamine (DA) in rat medial prefrontal cortex, as determined by in vivo microdialysis. Dextroamphetamine 118-139 5-hydroxytryptamine receptor 1A Rattus norvegicus 46-73 9113130-5 1996 Furthermore, not only elevated potassium ions, but D-amphetamine as well as GBR12909, all produced significant increases in the percentage spontaneous release of acetylcholinesterase. Dextroamphetamine 51-64 acetylcholinesterase Rattus norvegicus 162-182 8973993-12 1996 Serum prolactin levels were inversely associated with the amount of dextroamphetamine administered, with the largest decrease in serum prolactin levels observed after the 5-mg dose, and this finding was statistically significant. Dextroamphetamine 68-85 prolactin Homo sapiens 135-144 8973993-16 1996 Serum prolactin concentration was the most sensitive measure of central nervous system stimulation on EEG produced by dextroamphetamine under these study conditions. Dextroamphetamine 118-135 prolactin Homo sapiens 6-15 8933370-12 1996 Our findings demonstrate that 1) CGRP-LI can be measured in vivo in microdialysates from mPFC, vSTR, and hippocampus; 2) the release in vSTR is action potential-dependent; and 3) systemic administration of AMPH or PCP results in a long-lasting release of CGRP-LI in the mPFC and vSTR, thus demonstrating a novel action of these drugs in the brain. Dextroamphetamine 206-210 calcitonin-related polypeptide alpha Rattus norvegicus 33-37 8933370-12 1996 Our findings demonstrate that 1) CGRP-LI can be measured in vivo in microdialysates from mPFC, vSTR, and hippocampus; 2) the release in vSTR is action potential-dependent; and 3) systemic administration of AMPH or PCP results in a long-lasting release of CGRP-LI in the mPFC and vSTR, thus demonstrating a novel action of these drugs in the brain. Dextroamphetamine 206-210 calcitonin-related polypeptide alpha Rattus norvegicus 255-259 8959030-0 1996 Neurotoxicity of d-amphetamine in the C57BL/6J and CD-1 mouse. Dextroamphetamine 17-30 CD1 antigen complex Mus musculus 51-55 8880942-4 1996 On sample days, subjects were required to take d-amphetamine (10 mg BID) or placebo (0 mg BID) capsules. Dextroamphetamine 47-60 BH3 interacting domain death agonist Homo sapiens 68-71 7617701-5 1995 Pharmacological induction of increased locomotor activity as well as metabolic rate by d-amphetamine causes a significant increase in cardiac PBR binding, again, opposite to the response typically observed following inescapable shock stress. Dextroamphetamine 87-100 translocator protein Homo sapiens 142-145 8933370-3 1996 Consequently, we have here studied the effects of two psychotomimetic drugs, namely, d-amphetamine (AMPH) and phencyclidine (PCP), on CGRP concentrations in brain microdialysates from freely moving rats. Dextroamphetamine 85-98 calcitonin-related polypeptide alpha Rattus norvegicus 134-138 8933370-3 1996 Consequently, we have here studied the effects of two psychotomimetic drugs, namely, d-amphetamine (AMPH) and phencyclidine (PCP), on CGRP concentrations in brain microdialysates from freely moving rats. Dextroamphetamine 100-104 calcitonin-related polypeptide alpha Rattus norvegicus 134-138 8933370-9 1996 Both AMPH and PCP caused a significant and sustained increase (maximum about 300%) in CGRP-LI concentrations, in particular from the mPFC and vSTR, while saline had no effect. Dextroamphetamine 5-9 calcitonin-related polypeptide alpha Rattus norvegicus 86-90 8826542-1 1996 The study examined the effects of expectation on the subjective effects and oral self-administration of 15 mg d-amphetamine (AMP) and placebo in 40 volunteers who reported no prior use of stimulants other than caffeine. Dextroamphetamine 125-128 adenine phosphoribosyltransferase Homo sapiens 110-123 8723710-4 1996 D-Amphetamine, which releases DA from neurons and blocks the DA transporter directly, inhibited striatal [3H]WIN 35,428 binding in dose-dependent manner. Dextroamphetamine 0-13 solute carrier family 6 member 3 Homo sapiens 61-75 8782873-1 1996 Injections of D-amphetamine (5 mg/kg) three hours before sacrifice were found to elicit robust Fos-like immunoreactivity in the striatum of cynomolgus monkeys and of the common marmoset. Dextroamphetamine 14-27 proto-oncogene c-Fos Macaca fascicularis 95-98 8721156-3 1996 Twenty-four hours after lesion, both high (5 mg/kg) and low doses (0.5 mg/kg) of D-amphetamine induced contraversive rotation and intense striatal Fos activation on the denervated side; however, only the higher dose induced Fos on the normal side. Dextroamphetamine 81-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 147-150 8721156-3 1996 Twenty-four hours after lesion, both high (5 mg/kg) and low doses (0.5 mg/kg) of D-amphetamine induced contraversive rotation and intense striatal Fos activation on the denervated side; however, only the higher dose induced Fos on the normal side. Dextroamphetamine 81-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 224-227 7752096-1 1995 The dose-related effects of a single injection of D-amphetamine (AMPH) or methamphetamine (METH) on the mRNA expression of the immediate early gene zif/268 and the opioid peptide preprodynorphin, in rat forebrain, were investigated with quantitative in situ hybridization histochemistry 3 h after injection. Dextroamphetamine 50-63 early growth response 1 Rattus norvegicus 148-155 7752096-1 1995 The dose-related effects of a single injection of D-amphetamine (AMPH) or methamphetamine (METH) on the mRNA expression of the immediate early gene zif/268 and the opioid peptide preprodynorphin, in rat forebrain, were investigated with quantitative in situ hybridization histochemistry 3 h after injection. Dextroamphetamine 50-63 prodynorphin Rattus norvegicus 179-194 7752096-1 1995 The dose-related effects of a single injection of D-amphetamine (AMPH) or methamphetamine (METH) on the mRNA expression of the immediate early gene zif/268 and the opioid peptide preprodynorphin, in rat forebrain, were investigated with quantitative in situ hybridization histochemistry 3 h after injection. Dextroamphetamine 65-69 early growth response 1 Rattus norvegicus 148-155 7752096-1 1995 The dose-related effects of a single injection of D-amphetamine (AMPH) or methamphetamine (METH) on the mRNA expression of the immediate early gene zif/268 and the opioid peptide preprodynorphin, in rat forebrain, were investigated with quantitative in situ hybridization histochemistry 3 h after injection. Dextroamphetamine 65-69 prodynorphin Rattus norvegicus 179-194 7752096-4 1995 Dose-dependent induction of zif/268 mRNA also occurred in caudoputamen after injection of AMPH at all doses. Dextroamphetamine 90-94 early growth response 1 Rattus norvegicus 28-35 7752096-8 1995 These results demonstrate a clear dose-related responsiveness of zif/268 and preprodynorphin gene expression in cortical and/or striatal neurons that is positively correlated with a dose-dependent motor-stimulating effect of AMPH and METH. Dextroamphetamine 225-229 early growth response 1 Rattus norvegicus 65-72 7752096-8 1995 These results demonstrate a clear dose-related responsiveness of zif/268 and preprodynorphin gene expression in cortical and/or striatal neurons that is positively correlated with a dose-dependent motor-stimulating effect of AMPH and METH. Dextroamphetamine 225-229 prodynorphin Rattus norvegicus 77-92 8888359-6 1996 Administration of D-amphetamine increased plasma levels of cortisol, prolactin, growth hormone; elevated blood pressure, pulse, and temperature; and tended to increase self-ratings of activation/euphoria and anxiety. Dextroamphetamine 18-31 growth hormone 1 Homo sapiens 80-94 8783238-6 1996 Administration of d-amphetamine increased c-fos expression in the neostriatum and the globus pallidus of the control group. Dextroamphetamine 18-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 42-47 8783238-10 1996 Pre-treatment with SCH 23390 blocked the effect of d-amphetamine on c-fos expression in control and grafted animals. Dextroamphetamine 51-64 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 68-73 8558453-6 1996 Smaller doses of NMDAR1 antisense ODN did not lead to spontaneous rotation, but prominent ipsilateral rotation was observed after systemic administration of D-amphetamine. Dextroamphetamine 157-170 glutamate ionotropic receptor NMDA type subunit 1 Rattus norvegicus 17-23 8788863-1 1995 The effects of systemically administered phencyclidine (PCP; 2.5 mg/kg, s.c.) and D-amphetamine (1.5 mg/kg, s.c.) on the extracellular concentrations of neurotensin-like immunoreactivity (NT-LI) and dopamine (DA) in the ventral striatum (vSTR) and the medial prefrontal cortex (mPFC) were studied in freely moving rats using microdialysis. Dextroamphetamine 82-95 neurotensin Rattus norvegicus 153-164 7482290-3 1995 Fos immunoreactivity induced by d-amphetamine (5 mg/kg) or apomorphine (5 mg/kg) was quantified in dorsolateral and ventrolateral regions of caudate-putamen (CPu) in rats still demonstrating sensory neglect (5 days postsurgery) and in rats recovered from sensory neglect produced by AGm ablation (29+ days postsurgery). Dextroamphetamine 32-45 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 0-3 7482290-5 1995 In rats demonstrating sensory neglect, d-amphetamine or apomorphine induction of Fos in the ipsilateral CPu was reduced by about 40% compared to the contralateral CPu or to comparable readings in unlesioned controls. Dextroamphetamine 39-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 81-84 7753373-2 1995 Stimulants such as D-amphetamine and cocaine increase Fos-like immunoreactivity by enhancing the activation of D1 dopamine receptors. Dextroamphetamine 19-32 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 54-57 7753373-8 1995 Like the dorsal striatum, D1 agonists (D-amphetamine and CY 208-243), but not D2 antagonists (haloperidol and clozapine), increased Fos-like immunoreactivity in accumbal neurons projecting to the midbrain (ventral tegmental area and substantia nigra). Dextroamphetamine 39-52 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 132-135 7877442-6 1994 However, the AMPH-stimulated increase in PPD and zif/268 mRNA levels in striatum, but not zif/268 mRNA in cortex, was blocked by DNQX pretreatment. Dextroamphetamine 13-17 early growth response 1 Rattus norvegicus 49-56 7709341-0 1995 Cocaine and d-amphetamine increase c-fos expression in the rat cerebellum. Dextroamphetamine 12-25 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 35-40 7709341-2 1995 In the present study, immunohistochemical techniques were used to assess the pattern of c-fos expression in the cerebellum produced by d-amphetamine or cocaine. Dextroamphetamine 135-148 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 88-93 7709341-5 1995 Dose-dependent increases in Fos-like immunoreactivity were elicited by d-amphetamine and cocaine. Dextroamphetamine 71-84 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 28-31 7709341-8 1995 In addition, a homogeneous pattern of Fos-like immunoreactive nuclei, of sparse density, was also found near the pial surface of the molecular layer following d-amphetamine but not cocaine. Dextroamphetamine 159-172 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 38-41 7700517-2 1994 Using antisense oligonucleotides to c-fos, we demonstrate that D-amphetamine-induced c-fos expression can be attenuated in specific brain regions in vivo. Dextroamphetamine 63-76 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 36-41 7700517-2 1994 Using antisense oligonucleotides to c-fos, we demonstrate that D-amphetamine-induced c-fos expression can be attenuated in specific brain regions in vivo. Dextroamphetamine 63-76 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 85-90 7700517-3 1994 This unilateral attenuation of c-fos expression in D-amphetamine-stimulated animals results in a directed rotational behavior. Dextroamphetamine 51-64 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 31-36 7886627-1 1994 The role of N-methyl-D-aspartate (NMDA) excitatory amino acid receptors in D-amphetamine (AMPH)-induced behavioral changes and increased expression of the nuclear transcription factors, c-fos and zif/268, and preprodynorphin (PPD) mRNA in various regions of rat forebrain was investigated with quantitative in situ hybridization histochemistry. Dextroamphetamine 75-88 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 186-191 7886627-1 1994 The role of N-methyl-D-aspartate (NMDA) excitatory amino acid receptors in D-amphetamine (AMPH)-induced behavioral changes and increased expression of the nuclear transcription factors, c-fos and zif/268, and preprodynorphin (PPD) mRNA in various regions of rat forebrain was investigated with quantitative in situ hybridization histochemistry. Dextroamphetamine 75-88 early growth response 1 Rattus norvegicus 196-203 7886627-1 1994 The role of N-methyl-D-aspartate (NMDA) excitatory amino acid receptors in D-amphetamine (AMPH)-induced behavioral changes and increased expression of the nuclear transcription factors, c-fos and zif/268, and preprodynorphin (PPD) mRNA in various regions of rat forebrain was investigated with quantitative in situ hybridization histochemistry. Dextroamphetamine 75-88 prodynorphin Rattus norvegicus 209-224 7886627-1 1994 The role of N-methyl-D-aspartate (NMDA) excitatory amino acid receptors in D-amphetamine (AMPH)-induced behavioral changes and increased expression of the nuclear transcription factors, c-fos and zif/268, and preprodynorphin (PPD) mRNA in various regions of rat forebrain was investigated with quantitative in situ hybridization histochemistry. Dextroamphetamine 75-88 prodynorphin Rattus norvegicus 226-229 7886627-1 1994 The role of N-methyl-D-aspartate (NMDA) excitatory amino acid receptors in D-amphetamine (AMPH)-induced behavioral changes and increased expression of the nuclear transcription factors, c-fos and zif/268, and preprodynorphin (PPD) mRNA in various regions of rat forebrain was investigated with quantitative in situ hybridization histochemistry. Dextroamphetamine 90-94 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 186-191 7886627-1 1994 The role of N-methyl-D-aspartate (NMDA) excitatory amino acid receptors in D-amphetamine (AMPH)-induced behavioral changes and increased expression of the nuclear transcription factors, c-fos and zif/268, and preprodynorphin (PPD) mRNA in various regions of rat forebrain was investigated with quantitative in situ hybridization histochemistry. Dextroamphetamine 90-94 early growth response 1 Rattus norvegicus 196-203 7886627-5 1994 attenuated AMPH-induced striatal and cortical expression of zif/268 mRNA and striatal expression of PPD mRNA, without affecting the behavioral alterations induced by AMPH. Dextroamphetamine 11-15 early growth response 1 Rattus norvegicus 60-67 7886627-5 1994 attenuated AMPH-induced striatal and cortical expression of zif/268 mRNA and striatal expression of PPD mRNA, without affecting the behavioral alterations induced by AMPH. Dextroamphetamine 11-15 prodynorphin Rattus norvegicus 100-103 7886627-6 1994 A similar, dose-dependent suppression of AMPH-induced zif/268 and PPD mRNA in striatum and cortex was also revealed after systemic administration of (+/-)-3-(2-carboxypiperazin-4-yl)-propyl-1-phosphonic acid (CPP) at doses of 5 and 10 mg/kg. Dextroamphetamine 41-45 early growth response 1 Rattus norvegicus 54-61 7886627-6 1994 A similar, dose-dependent suppression of AMPH-induced zif/268 and PPD mRNA in striatum and cortex was also revealed after systemic administration of (+/-)-3-(2-carboxypiperazin-4-yl)-propyl-1-phosphonic acid (CPP) at doses of 5 and 10 mg/kg. Dextroamphetamine 41-45 prodynorphin Rattus norvegicus 66-69 7886627-10 1994 These studies indicate that NMDA receptors mediate, at least in part, activation of zif/268 and PPD gene expression in striatum and sensorimotor cortex by a single injection of AMPH. Dextroamphetamine 177-181 early growth response 1 Rattus norvegicus 84-91 7886627-10 1994 These studies indicate that NMDA receptors mediate, at least in part, activation of zif/268 and PPD gene expression in striatum and sensorimotor cortex by a single injection of AMPH. Dextroamphetamine 177-181 prodynorphin Rattus norvegicus 96-99 11224238-0 1994 Blockade of the discriminative stimulus effects of d-amphetamine in rhesus monkeys with serotonin 5-HT(1A) agonists. Dextroamphetamine 51-64 5-hydroxytryptamine receptor 1A Macaca mulatta 98-105 7821394-8 1994 A contraversive rotational bias was induced in BDNF and NT-4/5-treated rats challenged with d-amphetamine, and these responses were blocked by pretreatment with selective D1 or D2 receptor antagonists but not by opiate receptor antagonism. Dextroamphetamine 92-105 brain-derived neurotrophic factor Rattus norvegicus 47-51 7821394-8 1994 A contraversive rotational bias was induced in BDNF and NT-4/5-treated rats challenged with d-amphetamine, and these responses were blocked by pretreatment with selective D1 or D2 receptor antagonists but not by opiate receptor antagonism. Dextroamphetamine 92-105 neurotrophin 4 Rattus norvegicus 56-62 7821394-9 1994 Thus, NT-4/5 and BDNF can elevate the turnover of dopamine through both metabolic and release pools and augment the behavioral response to d-amphetamine. Dextroamphetamine 139-152 neurotrophin 4 Rattus norvegicus 6-12 7821394-9 1994 Thus, NT-4/5 and BDNF can elevate the turnover of dopamine through both metabolic and release pools and augment the behavioral response to d-amphetamine. Dextroamphetamine 139-152 brain-derived neurotrophic factor Rattus norvegicus 17-21 7969065-5 1994 Serotonin uptake and radioligand binding assays revealed that rat and human SERTs show different sensitivities to some but not all transporter ligands; most tricyclic antidepressants were significantly more potent at the human SERT, relative to rat SERT, whereas d-amphetamine was a more potent inhibitor of rat SERT. Dextroamphetamine 263-276 solute carrier family 6 member 4 Homo sapiens 76-80 7859063-4 1994 Studies on c-fos like immunoreactivity after systemic D-amphetamine treatment demonstrated a wide-spread appearance of c-fos like immunoreactive neuronal nuclear profiles within the neostriatum on both the unlesioned and denervated side. Dextroamphetamine 54-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 7859063-4 1994 Studies on c-fos like immunoreactivity after systemic D-amphetamine treatment demonstrated a wide-spread appearance of c-fos like immunoreactive neuronal nuclear profiles within the neostriatum on both the unlesioned and denervated side. Dextroamphetamine 54-67 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 119-124 11224238-9 1994 These results suggest that the discriminative stimulus effect of AMPH can be modulated by stimulation of 5-HT(1A) receptors to an extent comparable with that seen with a D2 antagonist. Dextroamphetamine 65-69 5-hydroxytryptamine receptor 1A Macaca mulatta 105-112 8042656-4 1994 D-amphetamine decreased aldosterone and renin levels in both groups. Dextroamphetamine 0-13 renin Homo sapiens 40-45 11224238-5 1994 Doses of the 5-HT(1A) agonists and raclopride, which occasioned only saline-lever responding when tested alone, shifted the AMPH dose-response function 0.5 to 1.0 log unit to the right in all monkeys, while pentobarbital had no effect. Dextroamphetamine 124-128 5-hydroxytryptamine receptor 1A Macaca mulatta 13-20 11224238-6 1994 For the 5-HT(1A) agonists, the order of potency for attenuating the AMPH discriminative stimulus was 8-OH-DPAT>buspirone>gepirone, similar to their binding affinities at 5-HT(1A) receptors. Dextroamphetamine 68-72 5-hydroxytryptamine receptor 1A Macaca mulatta 8-15 7948743-10 1994 Data from the present study show that IL-2 exerts the same neurochemical action as that previously observed with IFN-alpha for both d-amphetamine and EKC discrimination in rats. Dextroamphetamine 132-145 interleukin 2 Rattus norvegicus 38-42 8332620-0 1993 The selective 5-HT3 receptor antagonist, ondansetron, augments the anorectic effect of d-amphetamine in nondeprived rats. Dextroamphetamine 87-100 5-hydroxytryptamine receptor 3A Rattus norvegicus 14-28 7912402-4 1994 COS cell hSVMT expression yielded nanomolar affinities for tetrabenazine and reserpine, micromolar affinities for haloperidol, GBR12909, serotonin, mazindol, nomifensin and d-amphetamine, while dopamine, epinephrine, norepinephrine and histamine each displayed millimolar affinities. Dextroamphetamine 173-186 solute carrier family 18 member A2 Homo sapiens 9-14 7873126-1 1994 In the present study we estimated the effects of single and repeated administration of d-amphetamine (5 mg/kg, i.p., twice a day for 14 days) on tyrosine hydroxylase (TH) mRNA levels in the rat adrenal medulla. Dextroamphetamine 87-100 tyrosine hydroxylase Rattus norvegicus 145-165 7873126-1 1994 In the present study we estimated the effects of single and repeated administration of d-amphetamine (5 mg/kg, i.p., twice a day for 14 days) on tyrosine hydroxylase (TH) mRNA levels in the rat adrenal medulla. Dextroamphetamine 87-100 tyrosine hydroxylase Rattus norvegicus 167-169 7873126-2 1994 In situ hybridization experiments, conducted using a [35S]d-ATP-labelled deoxyoligonucleotide probe and a densitometric analysis of autoradiograms, showed that repeated d-amphetamine moderately increased the TH mRNA level (by ca. Dextroamphetamine 169-182 tyrosine hydroxylase Rattus norvegicus 208-210 7873126-7 1994 These results suggest that repeated d-amphetamine administration leads to biphasic changes in the adrenal TH biosynthesis, which may reflect an adaptive response to chronic drug treatment. Dextroamphetamine 36-49 tyrosine hydroxylase Rattus norvegicus 106-108 8473721-5 1993 d-Amphetamine produced a non-significant reduction in prolactin secretion. Dextroamphetamine 0-13 prolactin Homo sapiens 54-63 8298090-0 1993 Antisense oligonucleotide to c-fos induces ipsilateral rotational behaviour to d-amphetamine. Dextroamphetamine 79-92 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 29-34 8298090-3 1993 Here we can report that direct unilateral infusion into the rat neostriatum of an antisense phosphothioate oligodeoxynucleotide to c-fos mRNA leads to the rapid induction of ipsilateral rotational behaviour after d-amphetamine administration. Dextroamphetamine 213-226 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 131-136 8357529-9 1993 However, infusion of CCK immediately prior to D-amphetamine caused a dose-dependent potentiation of the impact of D-amphetamine upon rates of response on the CR lever. Dextroamphetamine 46-59 cholecystokinin Rattus norvegicus 21-24 8357529-9 1993 However, infusion of CCK immediately prior to D-amphetamine caused a dose-dependent potentiation of the impact of D-amphetamine upon rates of response on the CR lever. Dextroamphetamine 114-127 cholecystokinin Rattus norvegicus 21-24 8357529-13 1993 However, CCK again potentiated the D-amphetamine-induced increase in rates of response, and this potentiation was blocked by pretreatment with devazepide. Dextroamphetamine 35-48 cholecystokinin Rattus norvegicus 9-12 8473997-9 1993 l-Deprenyl produced clear generalization to the d-amphetamine stimulus only at very high doses of 17.0 to 30.0 mg/kg, doses about 10-fold higher than those that have a selective action on MAO-B vs. MAO-A and which start to have marked rate decreasing actions on food-reinforced responding. Dextroamphetamine 48-61 monoamine oxidase B Rattus norvegicus 188-193 7871033-2 1993 Seroquel and clozapine were differentially more active in reversing the inhibitory actions of d-amphetamine on mesolimbic (A10) than nigrostriatal (A9) dopamine (DA)-containing neurons, whereas haloperidol exhibited the opposite selectivity. Dextroamphetamine 94-107 immunoglobulin kappa variable 6D-21 (non-functional) Homo sapiens 123-126 8473721-0 1993 The prolactin response to d- and l-fenfluramine and to d-amphetamine in human subjects. Dextroamphetamine 55-68 prolactin Homo sapiens 4-13 8473721-1 1993 Twelve normal male volunteers took part in a double-blind placebo-controlled study to measure the effects of d- and l-fenfluramine singly and in combination with d-amphetamine on plasma prolactin levels. Dextroamphetamine 162-175 prolactin Homo sapiens 186-195 8097248-7 1993 This effect was mediated by SRIF1 receptors, as MK 678 (1-320 ng/side) produced a dose-dependent significant increase in locomotor activity with a maximal 228% increase relative to saline control, comparable to that attained with 3 to 10 micrograms of d-amphetamine. Dextroamphetamine 252-265 somatostatin receptor 2 Rattus norvegicus 28-33 8490714-0 1993 Central administration of corticotropin releasing factor induces long-term sensitization to D-amphetamine. Dextroamphetamine 92-105 corticotropin releasing hormone Homo sapiens 26-56 8453468-0 1993 Sensitization of c-fos expression in rat striatum following multiple challenges with D-amphetamine. Dextroamphetamine 85-98 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 17-22 8453468-1 1993 D-Amphetamine transiently stimulates the expression of the immediate-early response gene, c-fos, in rat striatal cell nuclei. Dextroamphetamine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 90-95 8453468-3 1993 induced a significantly greater expression of Fos-like immunoreactivity in striatum of rats treated three days previously with D-amphetamine compared to rats treated three days previously with saline. Dextroamphetamine 127-140 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 46-49 8453468-5 1993 This sensitization of c-fos expression following a repeated administration of D-amphetamine indicates an increased activation of post-synaptic elements in rat striatum. Dextroamphetamine 78-91 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 22-27 11224162-0 1992 Agonist and antagonist activity of low efficacy D2 dopamine receptor agonists in rats discriminating d-amphetamine from saline. Dextroamphetamine 101-114 dopamine receptor D2 Rattus norvegicus 48-68 11224165-1 1992 The effect of TRH pretreatment on locomotor activity and stereotypy induced by d-amphetamine was examined in rats. Dextroamphetamine 79-92 thyrotropin releasing hormone Rattus norvegicus 14-17 11224165-2 1992 The results show that TRH potentiates d-amphetamine-induced locomotor activity, but not d-amphetamine-induced stereotypy, suggesting that this neuropeptide selectively activates the mesolimbic dopamine system. Dextroamphetamine 38-51 thyrotropin releasing hormone Rattus norvegicus 22-25 1448483-9 1992 Both the NACT and NACP lesions attenuated the locomotor response to 1.5 mg/kg d-amphetamine sulphate IP, and the NACT group showed a supersensitive response to 0.1 mg/kg apomorphine HCl SC. Dextroamphetamine 78-100 solute carrier family 13 member 5 Rattus norvegicus 9-13 1933425-4 1991 Following the initial increase in DA caused by d-amphetamine, DA levels of CHP-treated rats were significantly lower than saline-treated rats across time (p less than 0.05). Dextroamphetamine 47-60 calcineurin-like EF-hand protein 1 Rattus norvegicus 75-78 1511338-3 1992 D-amphetamine and arecoline blocked the amnestic effect of beta-endorphin administered into the amygdala but it required higher doses for CCK-8, epinephrine and naloxone to block the amnestic effect of beta-endorphin. Dextroamphetamine 0-13 pro-opiomelanocortin-alpha Mus musculus 59-73 1511338-3 1992 D-amphetamine and arecoline blocked the amnestic effect of beta-endorphin administered into the amygdala but it required higher doses for CCK-8, epinephrine and naloxone to block the amnestic effect of beta-endorphin. Dextroamphetamine 0-13 pro-opiomelanocortin-alpha Mus musculus 202-216 1360906-4 1992 Administration of d-amphetamine induced an increase in the density of Fos-positive nuclei in the intact striatum. Dextroamphetamine 18-31 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 70-73 1740965-5 1992 The combination of the 5-HT-1a agonist 8-OH-DPAT and the 5-HT-1b agonist CGS-12066B, however, did inhibit d-amphetamine-stimulated locomotor activity. Dextroamphetamine 106-119 5-hydroxytryptamine receptor 1A Rattus norvegicus 23-30 1740965-5 1992 The combination of the 5-HT-1a agonist 8-OH-DPAT and the 5-HT-1b agonist CGS-12066B, however, did inhibit d-amphetamine-stimulated locomotor activity. Dextroamphetamine 106-119 5-hydroxytryptamine receptor 1B Rattus norvegicus 57-64 1683539-3 1991 In particular it has been shown that d-amphetamine can induce an activation of the c-fos early gene in various subregions of the neostriatum and that morphine treatment can antagonize this activation. Dextroamphetamine 37-50 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 1683539-1 1991 Focus on functional compartments produced by d-amphetamine activation of the c-fos gene and its relationship to the glucocorticoid receptor. Dextroamphetamine 45-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 77-82 1683539-1 1991 Focus on functional compartments produced by d-amphetamine activation of the c-fos gene and its relationship to the glucocorticoid receptor. Dextroamphetamine 45-58 nuclear receptor subfamily 3, group C, member 1 Rattus norvegicus 116-139 2021654-2 1991 In this study, with use of purified monoamine oxidase A, steady-state data for the inhibition by D-amphetamine of the oxidation of primary amines indicate the possibility of a ternary complex mechanism for monoamine oxidase A also. Dextroamphetamine 97-110 monoamine oxidase A Homo sapiens 36-55 2021654-2 1991 In this study, with use of purified monoamine oxidase A, steady-state data for the inhibition by D-amphetamine of the oxidation of primary amines indicate the possibility of a ternary complex mechanism for monoamine oxidase A also. Dextroamphetamine 97-110 monoamine oxidase A Homo sapiens 206-225 1672150-3 1991 In addition, whereas DA efflux was stimulated by either D-amphetamine (10 microM) or L-glutamate (10 mM) in the absence of Ca2+, these effects were greatly reduced when Mg2+ also was withdrawn from the buffer. Dextroamphetamine 56-69 mucin 7, secreted Homo sapiens 169-172 1679335-1 1991 d-Amphetamine (d-AMP) is a potent releaser of dopamine (DA), and its central nervous system stimulant action is mediated primarily through its effect on the substantia nigra and ventral tegmental area dopaminergic neurons (nuclei A9 and A10, respectively). Dextroamphetamine 15-20 UDP glucuronosyltransferase 1 family, polypeptide A7C Mus musculus 237-240 1679335-2 1991 The purpose of the present experiment was to use electrophysiological techniques to examine dendritic release of DA in the in vitro slice preparation, and determine whether: (1) d-AMP inhibits the firing rates of both A9 and A10 cells; (2) the d-AMP-induced inhibition is mediated via the dendritic release of DA; and (3) there is spontaneous dendritic release of DA. Dextroamphetamine 178-183 UDP glucuronosyltransferase 1 family, polypeptide A7C Mus musculus 225-228 1679335-3 1991 Superfusion with d-AMP (2-100 microM) produced identical inhibitory dose-response curves for A9 and A10 cells, and a dose of 6.25 microM caused more than 50% inhibition in the cell firing rates. Dextroamphetamine 17-22 UDP glucuronosyltransferase 1 family, polypeptide A7C Mus musculus 100-103 1679335-6 1991 These data suggest that d-AMP comparably releases DA from both A9 and A10 cell dendrites, that it releases newly-synthesized DA to inhibit cell firing, and that DA is tonically released to regulate cell firing rates via interactions with inhibitory D2 autoreceptors. Dextroamphetamine 24-29 UDP glucuronosyltransferase 1 family, polypeptide A7C Mus musculus 70-73 1646411-2 1991 This hypothesis has been tested here by investigating the effect of the met-enkephalin analog, DAMME (FK-33,824), on the elevation in serum cortisol induced by the catecholamine-releasing agent d-amphetamine (10 and 25 mg p.o.) Dextroamphetamine 194-207 proopiomelanocortin Homo sapiens 72-86 34906999-2 2022 Biochemical and crystallographic studies demonstrate that the competitive actions of DAT antagonists and substrates, such as cocaine and D-amphetamine (AMPH), rely on interactions with the transporter"s substrate binding site. Dextroamphetamine 137-150 Sodium-dependent dopamine transporter Caenorhabditis elegans 85-88 1679335-1 1991 d-Amphetamine (d-AMP) is a potent releaser of dopamine (DA), and its central nervous system stimulant action is mediated primarily through its effect on the substantia nigra and ventral tegmental area dopaminergic neurons (nuclei A9 and A10, respectively). Dextroamphetamine 0-13 UDP glucuronosyltransferase 1 family, polypeptide A7C Mus musculus 237-240 1977070-6 1990 Seven daily injections of AMP (5 mg/kg, Bid, s.c.), a DA-releasing agent, increased striatal DN-LI (187% of saline control) on the non-lesioned side, but not on the 6-OHDA-lesioned side. Dextroamphetamine 26-29 BH3 interacting domain death agonist Rattus norvegicus 40-43 1977070-8 1990 In contrast, [Met5]-enkephalin-like immunoreactivity (ME-LI) in the striatum was increased by a 6-OHDA-lesion (145% of contralateral control), which was blocked by repeated administration of APO but not AMP. Dextroamphetamine 203-206 proenkephalin Rattus norvegicus 20-30 34744015-5 2021 OXY-SAP-, but not BLANK-SAP- or PBS-treated rats, also showed marked dose-dependent rotational turning ipsilateral to the injected substantia nigra when challenged with d-amphetamine, but not with apomorphine. Dextroamphetamine 169-182 amyloid P component, serum Rattus norvegicus 4-7 34954300-0 2022 Differential Effects of Intra-Ventral Tegmental Area Ghrelin and Glucagon-like Peptide-1 on the Stimulatory Action of D-Amphetamine and Cocaine-Induced Ethanol Intake in Male Sprague Dawley Rats. Dextroamphetamine 118-131 ghrelin and obestatin prepropeptide Rattus norvegicus 53-60 34954300-7 2022 Our results indicated that VTA ghrelin significantly increased ethanol intake, and most importantly, potentiated the effect of d-amphetamine and cocaine on ethanol consumption. Dextroamphetamine 127-140 ghrelin and obestatin prepropeptide Rattus norvegicus 31-38 34364897-8 2021 Thus, after discontinuation of antipsychotic treatment, D1-and D2-mediated transmission differentially modulate the expression of a supersensitive response to d-amphetamine. Dextroamphetamine 159-172 solute carrier family 3 member 1 Rattus norvegicus 56-65 3174761-4 1988 ICV administered CCK-8 was found to antagonize the locomotor stimulatory effects of the low AMP dose, while the same peptide treatment markedly potentiated the stereotypy produced by the high dose of AMP. Dextroamphetamine 92-95 cholecystokinin Rattus norvegicus 17-20 35141357-4 2022 Cases: We report 6 cases with the KCNMA1-N999S variant treated with lisdexamfetamine (0.7-1.25 mg/kg/day), a pro-drug of dextroamphetamine. Dextroamphetamine 121-138 potassium calcium-activated channel subfamily M alpha 1 Homo sapiens 34-40 2532558-5 1989 D-Amphetamine induces both ipsilateral rotation and c-fos activation in the intact striatum. Dextroamphetamine 0-13 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 52-57 2625142-1 1989 Effect of some MAO inhibitors on rotational behavior induced by d-amphetamine in unilaterally 6-OHDA-lesioned rats was studied. Dextroamphetamine 64-77 monoamine oxidase A Rattus norvegicus 15-18 11175393-0 1989 Inhibitory effects of partial D2 dopamine receptor agonists on the d-amphetamine discriminative cue. Dextroamphetamine 67-80 dopamine receptor D2 Rattus norvegicus 30-50 2568305-0 1989 Pharmacological activity profiles of dopamine D-1 and D-2 reception agonists and antagonists on striatal neuronal activity and the response to dexamphetamine in freely moving rats. Dextroamphetamine 143-157 solute carrier family 3 member 1 Rattus norvegicus 37-57 35147850-7 2022 In nucleus accumbens, higher mRNA dopamine (D3) receptor expression was found in basal and D-amphetamine-challenge conditions in female than male, and prenatal amphetamine increased the difference. Dextroamphetamine 91-104 dopamine receptor D3 Rattus norvegicus 34-56 2502793-3 1989 Significant differences were found in (1) number of sessions to criterion (STC) (group B greater than group A); (2) group A Ss generalized both NTX and AMP to SAL, whereas group B Ss generalized AMP to the low dose (1.8 mg/kg) MS stimulus; and (3) in drug interaction test sessions, the high dose MS stimulus (10 mg/kg) in group A was unmodified by a range of challenge AMP doses (0.32 to 3.2 mg/kg). Dextroamphetamine 152-155 glycophorin A (MNS blood group) Homo sapiens 116-123 2502793-3 1989 Significant differences were found in (1) number of sessions to criterion (STC) (group B greater than group A); (2) group A Ss generalized both NTX and AMP to SAL, whereas group B Ss generalized AMP to the low dose (1.8 mg/kg) MS stimulus; and (3) in drug interaction test sessions, the high dose MS stimulus (10 mg/kg) in group A was unmodified by a range of challenge AMP doses (0.32 to 3.2 mg/kg). Dextroamphetamine 152-155 glycophorin A (MNS blood group) Homo sapiens 116-123 3174761-4 1988 ICV administered CCK-8 was found to antagonize the locomotor stimulatory effects of the low AMP dose, while the same peptide treatment markedly potentiated the stereotypy produced by the high dose of AMP. Dextroamphetamine 200-203 cholecystokinin Rattus norvegicus 17-20 2835690-5 1988 Inhibition of type A MAO with clorgyline (0.1 mumol/l) significantly enhanced the increase in cyclic AMP formation seen after d-amphetamine. Dextroamphetamine 126-139 monoamine oxidase A Rattus norvegicus 21-24 2896544-0 1988 Long term chronic D-amphetamine-induced histobiochemical alterations of GD and AAT enzymatic activities in the rat cerebellum. Dextroamphetamine 18-31 serpin family A member 1 Rattus norvegicus 79-82 3320348-5 1987 Nonopioid compounds, such as d-amphetamine, pentobarbital, phencyclidine and (+)-SKF 10,047 produced responding predominantly on the water-appropriate key in both U50,488- and morphine-trained pigeons. Dextroamphetamine 29-42 small nucleolar RNA, C/D box 50B Homo sapiens 163-166 2430671-2 1986 SP immunoneutralization in the NAS also reduced the locomotor response to systemically administered D-amphetamine. Dextroamphetamine 100-113 tachykinin precursor 1 Homo sapiens 0-2 3037425-7 1987 Citrate synthase was the only enzyme in the Krebs" cycle affected by chronic administration of dextroamphetamine. Dextroamphetamine 95-112 citrate synthase Rattus norvegicus 0-16 3659117-2 1987 significantly (P less than 0.05) decreased the rate of CAR in the shuttle box avoidance test and antagonized the d-amphetamine (d-A) (1 mg/kg s.c.)-induced increase of CAR. Dextroamphetamine 113-126 nuclear receptor subfamily 1, group I, member 3 Rattus norvegicus 168-171 3659117-2 1987 significantly (P less than 0.05) decreased the rate of CAR in the shuttle box avoidance test and antagonized the d-amphetamine (d-A) (1 mg/kg s.c.)-induced increase of CAR. Dextroamphetamine 128-131 nuclear receptor subfamily 1, group I, member 3 Rattus norvegicus 168-171 2950291-7 1987 The marked potency of SCH 23390 in reversing d-amphetamine could be due to its combined antagonist effects at 5HT2 and D-1 DA receptor sites. Dextroamphetamine 45-58 leiomodin 1 Homo sapiens 119-122 3019477-8 1986 D-(+)-Amphetamine (1 microM), when superfused in the CN, induced a large enhancement of the [3H]DA release in the ipsilateral ACE simultaneously with the local increase of [3H]DA release. Dextroamphetamine 0-17 angiotensin I converting enzyme Rattus norvegicus 126-129 3748452-0 1986 Repeated neurotensin administration in the ventral tegmental area: effects on baseline and D-amphetamine-induced locomotor activity. Dextroamphetamine 91-104 neurotensin Rattus norvegicus 9-20 3748325-0 1986 Centrally administered neurotensin suppresses locomotor hyperactivity induced by d-amphetamine but not by scopolamine or caffeine. Dextroamphetamine 81-94 neurotensin Rattus norvegicus 23-34 3748325-8 1986 Statistical analysis revealed that neurotensin significantly suppressed the locomotor response to 2 and 3 mg/kg of d-amphetamine but did not suppress the locomotor responses to any dose of scopolamine or caffeine or the non-locomotor responses to any of the three stimulants tested. Dextroamphetamine 115-128 neurotensin Rattus norvegicus 35-46 3708329-4 1986 On the other hand, preincubation of striatal slices with D-amphetamine (10(-5) M) enhanced basal and veratrine-stimulated dopamine release but markedly suppressed evoked CCK release. Dextroamphetamine 57-70 cholecystokinin Rattus norvegicus 170-173 3841736-1 1985 Acute injection of d-amphetamine (10 mg/kg), administered to rats 60 minutes prior to sacrifice, induced a doubling of immunoreactive NPY (NPY-IR) in pineal gland. Dextroamphetamine 19-32 neuropeptide Y Rattus norvegicus 134-137 3841736-1 1985 Acute injection of d-amphetamine (10 mg/kg), administered to rats 60 minutes prior to sacrifice, induced a doubling of immunoreactive NPY (NPY-IR) in pineal gland. Dextroamphetamine 19-32 neuropeptide Y Rattus norvegicus 139-145 6698380-0 1984 Sex-related differences in pineal gland N-acetyltransferase induction by d-amphetamine. Dextroamphetamine 73-86 N-acetyltransferase 1 Rattus norvegicus 40-59 2932759-0 1985 Different pattern of association of beta-endorphin and cortisol responses to dextroamphetamine in postmenopausal women and young men. Dextroamphetamine 77-94 proopiomelanocortin Homo sapiens 36-50 2932759-1 1985 A negative correlation between plasma beta-endorphin and cortisol responses to 0.15 mg/kg dextroamphetamine i.v. Dextroamphetamine 90-107 proopiomelanocortin Homo sapiens 38-52 2863810-0 1985 The effects of MIF-I, beta-endorphin and alpha-MSH on d-amphetamine induced paradoxical behavioral thermoregulation: possible involvement of the dopaminergic system. Dextroamphetamine 54-67 proopiomelanocortin Rattus norvegicus 41-50 2997819-1 1985 We report the effects of intracerebroventricular (ICV) administration of thyrotropin releasing hormone (TRH), a TRH metabolite histidyl-proline diketopiperazine (DKP) and systemically administered d-amphetamine (AMP) on the locomotor activity of two groups of rats which had previously received bilateral injections of either 6-hydroxydopamine (6-OHDA) or saline into the nucleus accumbens. Dextroamphetamine 197-210 thyrotropin releasing hormone Rattus norvegicus 104-107 2997819-1 1985 We report the effects of intracerebroventricular (ICV) administration of thyrotropin releasing hormone (TRH), a TRH metabolite histidyl-proline diketopiperazine (DKP) and systemically administered d-amphetamine (AMP) on the locomotor activity of two groups of rats which had previously received bilateral injections of either 6-hydroxydopamine (6-OHDA) or saline into the nucleus accumbens. Dextroamphetamine 212-215 thyrotropin releasing hormone Rattus norvegicus 73-102 2997819-1 1985 We report the effects of intracerebroventricular (ICV) administration of thyrotropin releasing hormone (TRH), a TRH metabolite histidyl-proline diketopiperazine (DKP) and systemically administered d-amphetamine (AMP) on the locomotor activity of two groups of rats which had previously received bilateral injections of either 6-hydroxydopamine (6-OHDA) or saline into the nucleus accumbens. Dextroamphetamine 212-215 thyrotropin releasing hormone Rattus norvegicus 104-107 2997819-5 1985 However, coadministration of TRH or DKP with systemically administered AMP enhanced rotation above that found after injection of AMP alone. Dextroamphetamine 71-74 thyrotropin releasing hormone Rattus norvegicus 29-32 2997819-5 1985 However, coadministration of TRH or DKP with systemically administered AMP enhanced rotation above that found after injection of AMP alone. Dextroamphetamine 129-132 thyrotropin releasing hormone Rattus norvegicus 29-32 4005584-10 1985 D-amphetamine was more effective than L-amphetamine in increasing peak 1 height. Dextroamphetamine 0-13 pseudopodium-enriched atypical kinase 1 Rattus norvegicus 66-72 2992996-3 1985 The effects of TRH-A (10(-4)M) were abolished by 10(-7) M TRH, but only partially (about 50%) inhibited by 10(-4) M d-amphetamine. Dextroamphetamine 116-129 thyrotropin releasing hormone Cavia porcellus 15-18 2992996-5 1985 However, the duodenal response to TRH (10(-6) M) was dose dependently inhibited by d-amphetamine (10(-6), 10(-5), 10(-4) M) while the phasic and tonic contractions caused by high K+ (40 mM) or the contractile responses to acetylcholine (10(-7) M) were not blocked by d-amphetamine. Dextroamphetamine 83-96 thyrotropin releasing hormone Cavia porcellus 34-37 2992996-5 1985 However, the duodenal response to TRH (10(-6) M) was dose dependently inhibited by d-amphetamine (10(-6), 10(-5), 10(-4) M) while the phasic and tonic contractions caused by high K+ (40 mM) or the contractile responses to acetylcholine (10(-7) M) were not blocked by d-amphetamine. Dextroamphetamine 267-280 thyrotropin releasing hormone Cavia porcellus 34-37 2992996-6 1985 These results indicate that d-amphetamine may act as an antagonist to TRH without influencing the movement of calcium ions in smooth muscle or muscarinic receptors and that contractile responses to TRH-A are mediated through TRH receptors in the myenteric cholinergic nerves. Dextroamphetamine 28-41 thyrotropin releasing hormone Cavia porcellus 70-73 6149772-7 1984 PRL decreased from baseline following both d-amphetamine and placebo and there was no significant drug-placebo difference. Dextroamphetamine 43-56 prolactin Homo sapiens 0-3 6201607-4 1984 Subacute treatment with d-amphetamine exerted METH-like effects on the substance P neurons of the basal ganglia, whereas haloperidol completely antagonized the METH effect. Dextroamphetamine 24-37 tachykinin precursor 1 Homo sapiens 71-82 6708722-1 1984 The present experiments investigated the relationship between the spontaneous basal firing rate of A10 dopamine (DA) neurons and their sensitivity to the rate-suppressant effects of intravenously administered apomorphine (APO) and d-amphetamine (AMP) as well as microiontophoretically ejected DA. Dextroamphetamine 231-244 immunoglobulin kappa variable 6D-21 (non-functional) Homo sapiens 99-102 6436877-9 1984 d-Amphetamine sulphate (0.25, 0.75 mg/kg SC salt), given 15 min before testing, also increased SST and stimulated responding. Dextroamphetamine 0-22 somatostatin Rattus norvegicus 95-98 6651958-2 1983 Consistent with previous data, tone preexposure resulted in retarded acquisition of the conditioned avoidance response (CAR) in saline control animals and in animals that received chronic administration of a low dose of d-amphetamine. Dextroamphetamine 220-233 nuclear receptor subfamily 1, group I, member 3 Rattus norvegicus 120-123 6124951-0 1982 Modification of d-amphetamine- or chlorpromazine-induced hypothermia by beta-endorphin, MIF-I, and alpha-MSH: mediation by the dopaminergic system. Dextroamphetamine 16-29 macrophage migration inhibitory factor Rattus norvegicus 88-91 6642414-1 1983 The effect on plasma prolactin (PRL) of d-amphetamine (Amph) was studied in normo- and hyperprolactinemic subjects. Dextroamphetamine 40-53 prolactin Homo sapiens 21-30 6642414-1 1983 The effect on plasma prolactin (PRL) of d-amphetamine (Amph) was studied in normo- and hyperprolactinemic subjects. Dextroamphetamine 40-53 prolactin Homo sapiens 32-35 6642414-1 1983 The effect on plasma prolactin (PRL) of d-amphetamine (Amph) was studied in normo- and hyperprolactinemic subjects. Dextroamphetamine 55-59 prolactin Homo sapiens 21-30 6642414-1 1983 The effect on plasma prolactin (PRL) of d-amphetamine (Amph) was studied in normo- and hyperprolactinemic subjects. Dextroamphetamine 55-59 prolactin Homo sapiens 32-35 6642414-2 1983 In normoprolactinemic women Amph failed to lower plasma PRL levels when infused intravenously over 1 h at the dose of 7.5 mg, but induced at the dose of 15.0 mg a modest inhibition of plasma PRL (maximum PRL inhibition 20 +/- 4.5% at 45 min). Dextroamphetamine 28-32 prolactin Homo sapiens 191-194 6642414-2 1983 In normoprolactinemic women Amph failed to lower plasma PRL levels when infused intravenously over 1 h at the dose of 7.5 mg, but induced at the dose of 15.0 mg a modest inhibition of plasma PRL (maximum PRL inhibition 20 +/- 4.5% at 45 min). Dextroamphetamine 28-32 prolactin Homo sapiens 191-194 6642414-5 1983 These results indicate that Amph is a poor PRL suppressor in either normo- or hyperprolactinemic subjects. Dextroamphetamine 28-32 prolactin Homo sapiens 43-46 6356168-1 1983 The influence of luteinizing hormone releasing hormone (LHRH) on the behavioral effects induced by several doses of D-amphetamine (0.25, 0.5, 1.0 and 2.0 mg/kg IP) was studied. Dextroamphetamine 116-129 gonadotropin releasing hormone 1 Rattus norvegicus 56-60 6356168-4 1983 Pretreatment with 100 micrograms/kg of LHRH antagonizes the enhancement in acquisition of CARs due to D-amphetamine 0.5, 1.0 and 2.0 mg/kg, the impairment in retention induced by amphetamine 1.0 and 2.0 mg/kg, and the hypermotility and the increased rearing behavior induced by amphetamine 1.0 and 2.0 mg/kg. Dextroamphetamine 102-115 gonadotropin releasing hormone 1 Rattus norvegicus 39-43 6682440-2 1983 injection of neurotensin (NT) to rats or mice attenuated the locomotor hyperactivity induced by d-amphetamine, methylphenidate or cocaine, but not the increased activity induced by apomorphine or lergotrile. Dextroamphetamine 96-109 neurotensin Rattus norvegicus 13-24 6652649-0 1983 Action of D-amphetamine on the GABA-T histochemical reaction in several nervous centers. Dextroamphetamine 10-23 4-aminobutyrate aminotransferase Homo sapiens 31-37 6292981-0 1982 Response of plasma beta-endorphin immunoreactivity to d-amphetamine and placebo in schizophrenic patients. Dextroamphetamine 54-67 proopiomelanocortin Homo sapiens 19-33 6292981-1 1982 The response of plasma beta-endorphin (ir) to infusions of randomly assigned d-amphetamine (20 mg) and placebo was studied in eight schizophrenic patients. Dextroamphetamine 77-90 proopiomelanocortin Homo sapiens 23-37 7115417-6 1982 In scRes monoamine oxidase (MAO)-inhibited preparations, Amph released preloaded [3H]DA located in the cytosol in the absence of functional vesicles. Dextroamphetamine 57-61 monoamine oxidase A Rattus norvegicus 9-26 7115417-6 1982 In scRes monoamine oxidase (MAO)-inhibited preparations, Amph released preloaded [3H]DA located in the cytosol in the absence of functional vesicles. Dextroamphetamine 57-61 monoamine oxidase A Rattus norvegicus 28-31 6124951-0 1982 Modification of d-amphetamine- or chlorpromazine-induced hypothermia by beta-endorphin, MIF-I, and alpha-MSH: mediation by the dopaminergic system. Dextroamphetamine 16-29 proopiomelanocortin Rattus norvegicus 99-108 6124951-1 1982 The effects of beta-endorphin, MIF-I, and alpha-MSH on d-amphetamine- a CPZ-induced hypothermias in rats kept at 4 degrees C were tested in three experimental groups: (a) intact; (b) rats with lesions of the olfactory tubercle; and (c) rats in which the link between the DA mesolimbic pathway and the striatum was disconnected. Dextroamphetamine 55-68 carboxypeptidase Z Rattus norvegicus 72-75 6124951-3 1982 Pretreatment with CPZ, MIF-I, and alpha-MSH potentiated d-amphetamine-induced hypothermia in intact rats. Dextroamphetamine 56-69 carboxypeptidase Z Rattus norvegicus 18-21 6124951-3 1982 Pretreatment with CPZ, MIF-I, and alpha-MSH potentiated d-amphetamine-induced hypothermia in intact rats. Dextroamphetamine 56-69 macrophage migration inhibitory factor Rattus norvegicus 23-26 6124951-3 1982 Pretreatment with CPZ, MIF-I, and alpha-MSH potentiated d-amphetamine-induced hypothermia in intact rats. Dextroamphetamine 56-69 proopiomelanocortin Rattus norvegicus 34-43 7065832-0 1982 Growth hormone response to dextroamphetamine in depressed patients and normal subjects. Dextroamphetamine 27-44 growth hormone 1 Homo sapiens 0-14 7065832-1 1982 The human growth hormone (HGH) response to dextroamphetamine sulfate (doses, 0.1 and 0.15 mg/kg) was determined in both the morning and evening in patients with endogenous and atypical depression and in normal young men and normal postmenopausal women. Dextroamphetamine 43-68 growth hormone 1 Homo sapiens 10-24 7155357-2 1982 Direct infusion of d-amphetamine (10(-6) M) to one substantia nigra evoked a release of acetylcholinesterase and aminopeptidase not only locally, but also from the contralateral caudate nucleus and substantia nigra. Dextroamphetamine 19-32 ACE-1 Oryctolagus cuniculus 88-108 7200582-4 1981 d-Amphetamine non-competitively inhibited rat eye ALDH in vitro. Dextroamphetamine 0-13 aldehyde dehydrogenase 3 family, member A1 Rattus norvegicus 50-54 6272039-0 1981 Dextroamphetamine infusions in normals result in correlated increases of plasma beta-endorphin and cortisol immunoreactivity. Dextroamphetamine 0-17 proopiomelanocortin Homo sapiens 80-94 6794010-5 1981 While TRH partially blocked d-amphetamine-induced hypothermia were blocked in olfactory tubercle-lesioned rats. Dextroamphetamine 28-41 thyrotropin releasing hormone Rattus norvegicus 6-9 7253823-0 1981 The prolactin response to intravenous dextroamphetamine in normal young men and postmenopausal women. Dextroamphetamine 38-55 prolactin Homo sapiens 4-13 45570-2 1979 AHR-6646 blocked d-amphetamine lethality in mice under aggregated conditions when the pretreatment interval was between one hour and seven days. Dextroamphetamine 17-30 aryl-hydrocarbon receptor Mus musculus 0-3 7348023-3 1981 Aminooxyacetic acid (12.5 mg/kg) as a GABA-transaminase inhibitor, decreased the rotation elicited by d-amphetamine or apomorphine. Dextroamphetamine 102-115 4-aminobutyrate aminotransferase Rattus norvegicus 38-55 7217554-0 1981 Growth hormone, prolactin, and growth responses in hyperkinetic males treated with d-amphetamine. Dextroamphetamine 83-96 growth hormone 1 Homo sapiens 0-14 7193892-0 1980 Diurnal growth hormone responses to dextroamphetamine in normal young men and post-menopausal women. Dextroamphetamine 36-53 growth hormone 1 Homo sapiens 8-22 40668-0 1979 Nerve growth factor: effects on D-amphetamine-induced activity and brain monoamines. Dextroamphetamine 32-45 nerve growth factor Rattus norvegicus 0-19 40668-4 1979 NGF-treated rats showed an enhanced response to D-amphetamine (1.5 mg/kg) when tested 15 days postoperatively. Dextroamphetamine 48-61 nerve growth factor Rattus norvegicus 0-3 40668-6 1979 Intracerebral NGF administeration enhanced the response to D-amphetamine 15 days later in rats without lesions, and also appeared to result in increased turnover of brain norepinephrine and serotonin at 3, but not 15, days postadministration. Dextroamphetamine 59-72 nerve growth factor Rattus norvegicus 14-17 288372-3 1979 Dopaminergic agonists (apomorphine, piribedil, d-amphetamine, L-DOPA, and the ergot derivatives bromocriptine and lisuride) all caused a decrease of serum prolactin levels. Dextroamphetamine 47-60 prolactin Homo sapiens 155-164 438802-1 1979 The effect of d-amphetamine on in vivo catecholamine synthesis in four regions of rat brain was determined by measuring the accumulation of dopa after inhibiton of dopa decarboxylase. Dextroamphetamine 14-27 dopa decarboxylase Rattus norvegicus 164-182 745694-0 1978 The effect of oral dextroamphetamine on prolactin secretion in man. Dextroamphetamine 19-36 prolactin Homo sapiens 40-49 318182-2 1978 d-Amphetamine, 10(-4)M, noncompetitively inhibited rat and mouse L-ALDH in vitro. Dextroamphetamine 0-13 aldehyde dehydrogenase family 3, subfamily A1 Mus musculus 67-71 362103-0 1978 d-Amphetamine raises serum prolactin in man: evaluations after chronic placebo, lithium and pimozide treatment. Dextroamphetamine 0-13 prolactin Homo sapiens 27-36 660556-7 1978 Reserpine and d-amphetamine in vivo elicited an increase and decrease, respectively, in striatal PH4 paralleling induced changes in synaptosomal DA synthesis. Dextroamphetamine 14-27 prolyl 4-hydroxylase, transmembrane Rattus norvegicus 97-100 674916-0 1978 [Increased somatotrophin response to insulin induced hypoglycemia with D-amphetamine treatment (author"s transl)]. Dextroamphetamine 71-84 insulin Homo sapiens 37-44 414276-0 1977 Blockage of progesterone-induced release of luteinizing hormone and prolactin by d-amphetamine and fenfluramine in rats. Dextroamphetamine 81-94 prolactin Rattus norvegicus 68-77 414276-2 1977 In these animals prolactin levels decreased after injection of 0.6 and 1.25 mg/kg of d-amphetamine. Dextroamphetamine 85-98 prolactin Rattus norvegicus 17-26 414276-6 1977 It is concluded that d-amphetamine and fenfluramine are able to alter the facilitatory actions of progesterone on luteinizing hormone and prolactin release in ovariectomized estradiol-primed rats. Dextroamphetamine 21-34 prolactin Rattus norvegicus 138-147 33826955-4 2021 Male C57BL/6 mice showed increased c-FOS expression in PAG dopamine neurons and a significant increase in paw withdrawal latency to thermal stimulation after receiving a systemic injection of D-amphetamine. Dextroamphetamine 192-205 FBJ osteosarcoma oncogene Mus musculus 35-40 11829024-12 1977 Scopolamine/Dexedrine caused consistent elevations in urinary cortisol and epinephrine and a transient elevation in ADH. Dextroamphetamine 12-21 arginine vasopressin Homo sapiens 116-119 318680-0 1977 The effect of the stimulant drugs, dextroamphetamine and methylphenidate, on secretion of growth hormone in hyperactive children. Dextroamphetamine 35-52 growth hormone 1 Homo sapiens 90-104 318680-5 1977 The findings indicate an acute and a probably long-term effect of dextroamphetamine and methylphenidate on the homeostasis of growth hormone. Dextroamphetamine 66-83 growth hormone 1 Homo sapiens 126-140 1028957-2 1976 In contrast, the dopaminergic agonists apomorphine (APO), lisuride hydrogen meleate (LHM), D-amphetamine (AMPH), piribedil and L-dopa greatly lowered the high serum Prl concentrations in female rats induced by i.p. Dextroamphetamine 106-110 prolactin Rattus norvegicus 165-168 19604784-0 1975 Growth hormone response to D-amphetamine in normal controls and in depressive patients. Dextroamphetamine 27-40 growth hormone 1 Homo sapiens 0-14 4154853-0 1974 Blockade by prostaglandins E2 and F1 alpha of the response of the rabbit ileum to stimulation of sympathetic nerve and its reversal by some antihistamines, dexamphetamine and methylphenidate. Dextroamphetamine 156-170 E2 small nucleolar RNA Oryctolagus cuniculus 27-42 1252961-1 1976 The direct application of crystalline dopamine, D-amphetamine or scopolamine in microgram quantities to the ventral anterior region of the corpus striatum (VAS) of rats increased their responding for food on a modified DRL-30 sec schedule of reinforcement. Dextroamphetamine 48-61 arginine vasopressin Rattus norvegicus 156-159 178070-0 1976 Effects of imipramine, nomifensine and d-amphetamine on type B monoamine oxidase, cyclic AMP phosphodiesterase and dopamine-beta-hydroxylase. Dextroamphetamine 39-52 dopamine beta-hydroxylase Homo sapiens 115-140 178070-1 1976 The three dissimilar structural antidepressant drugs, imipramine, nomifensine and d-amphetamine, were compared for their effects on type B monoamine oxidase, cyclic AMP phosphodiesterase and dopamine-beta-hydroxylase. Dextroamphetamine 82-95 dopamine beta-hydroxylase Homo sapiens 191-216 1171512-1 1975 Chlorphentermine, fenfluramine and dexamphetamine have a pronounced stimulating effect on lipolysis as determined by the action of mouse pancreas lipase on the dilauric (didodecanoic) acid ester of fluorescein. Dextroamphetamine 35-49 lipase, endothelial Mus musculus 146-152 33976392-4 2021 Compared to wildtype controls, the Grk3-/- mice show a number of aberrations linked to psychosis, including elevated brain levels of IL-1beta, increased turnover of kynurenic acid (KYNA), hyper-responsiveness to D-amphetamine, elevated spontaneous firing of midbrain dopamine neurons, and disruption in prepulse inhibition. Dextroamphetamine 212-225 G protein-coupled receptor kinase 3 Mus musculus 35-39 33908633-0 2021 Quercetin Relieves D-Amphetamine induced Manic-like Behavior through Activating TREK-1 Potassium Channels in Mice. Dextroamphetamine 19-32 potassium channel, subfamily K, member 2 Mus musculus 80-86 33191076-11 2021 DOXY, better than lithium, reversed the d-amphetamine-induced rise in TNFalpha, MPO, and lipid peroxidation. Dextroamphetamine 40-53 tumor necrosis factor Mus musculus 70-78 33191076-11 2021 DOXY, better than lithium, reversed the d-amphetamine-induced rise in TNFalpha, MPO, and lipid peroxidation. Dextroamphetamine 40-53 myeloperoxidase Mus musculus 80-83 32682325-2 2021 Preclinical studies using Long Access (LgA) cocaine self-administration procedures suggest D-amphetamine may act by preventing tolerance to cocaine"s effects at the dopamine transporter (DAT). Dextroamphetamine 91-104 solute carrier family 6 member 3 Rattus norvegicus 165-185 32682325-2 2021 Preclinical studies using Long Access (LgA) cocaine self-administration procedures suggest D-amphetamine may act by preventing tolerance to cocaine"s effects at the dopamine transporter (DAT). Dextroamphetamine 91-104 solute carrier family 6 member 3 Rattus norvegicus 187-190 32859997-0 2021 D-amphetamine maintenance treatment goes a long way: lasting therapeutic effects on cocaine behavioral effects and cocaine potency at the dopamine transporter. Dextroamphetamine 0-13 solute carrier family 6 member 3 Homo sapiens 138-158 32682325-10 2021 After cessation of D-amphetamine treatment, the motivation to take and seek cocaine was also reduced, and sensitization of cocaine"s actions at the DAT was reversed. Dextroamphetamine 19-32 solute carrier family 6 member 3 Rattus norvegicus 148-151 32682325-11 2021 Thus, treatment with D-amphetamine might reduce cocaine use by preventing sensitization-related changes in cocaine potency at the DAT, consistent with an incentive-sensitization view of addiction. Dextroamphetamine 21-34 solute carrier family 6 member 3 Rattus norvegicus 130-133 31883280-0 2020 Lithium attenuates d-amphetamine-induced hyperlocomotor activity in mice via inhibition of interaction between cyclooxygenase-2 and indoleamine-2,3-dioxygenase. Dextroamphetamine 19-32 prostaglandin-endoperoxide synthase 2 Mus musculus 111-127 33368923-0 2021 Regulation of GluA1 phosphorylation by d-amphetamine and methylphenidate in the cerebellum. Dextroamphetamine 39-52 glutamate receptor, ionotropic, AMPA1 (alpha 1) Mus musculus 14-19 33368923-4 2021 Here, we demonstrate that d-amphetamine and methylphenidate increase phosphorylation at Ser845 (pS845-GluA1) in the membrane fraction of mouse cerebellum homogenate. Dextroamphetamine 26-39 glutamate receptor, ionotropic, AMPA1 (alpha 1) Mus musculus 102-107 33368923-6 2021 Consequently, d-amphetamine-induced pS845-GluA1 was prevented by beta1-adenoreceptor antagonist, whereas the blockade of DA D1 receptor had no effect. Dextroamphetamine 14-27 glutamate receptor, ionotropic, AMPA1 (alpha 1) Mus musculus 42-47 32165217-0 2020 NQO1 regulates pharmaco-behavioral effects of d-amphetamine in striatal dopaminergic system in mice. Dextroamphetamine 46-59 NAD(P)H dehydrogenase, quinone 1 Mus musculus 0-4 32165217-5 2020 According to our comparative study involving NQO1+/+ and NQO1-/- mice, NQO1 deficiency increased d-amphetamine-induced psychomotor activity and psychological dependency compared to wild-type mice. Dextroamphetamine 97-110 NAD(P)H dehydrogenase, quinone 1 Mus musculus 57-61 32165217-5 2020 According to our comparative study involving NQO1+/+ and NQO1-/- mice, NQO1 deficiency increased d-amphetamine-induced psychomotor activity and psychological dependency compared to wild-type mice. Dextroamphetamine 97-110 NAD(P)H dehydrogenase, quinone 1 Mus musculus 57-61 32165217-6 2020 Basal and d-amphetamine-induced dopamine levels were also enhanced by NQO1 deficiency. Dextroamphetamine 10-23 NAD(P)H dehydrogenase, quinone 1 Mus musculus 70-74 32165217-7 2020 In NQO1-/- mice, neural activation induced by d-amphetamine was higher in dorsolateral striatum, but not in dorsomedial and ventral striata. Dextroamphetamine 46-59 NAD(P)H dehydrogenase, quinone 1 Mus musculus 3-7 31883280-2 2020 Repeated treatment with AMP resulted in significant increases in proinflammatory cyclooxygenase-2 (COX-2) and indolemaine-2,3-dioxygenase-1 (IDO)-1 expression in the prefrontal cortex (PFC) of mice. Dextroamphetamine 24-27 prostaglandin-endoperoxide synthase 2 Mus musculus 81-97 31883280-2 2020 Repeated treatment with AMP resulted in significant increases in proinflammatory cyclooxygenase-2 (COX-2) and indolemaine-2,3-dioxygenase-1 (IDO)-1 expression in the prefrontal cortex (PFC) of mice. Dextroamphetamine 24-27 prostaglandin-endoperoxide synthase 2 Mus musculus 99-104 31883280-2 2020 Repeated treatment with AMP resulted in significant increases in proinflammatory cyclooxygenase-2 (COX-2) and indolemaine-2,3-dioxygenase-1 (IDO)-1 expression in the prefrontal cortex (PFC) of mice. Dextroamphetamine 24-27 indoleamine 2,3-dioxygenase 1 Mus musculus 110-147 31883280-3 2020 However, AMP treatment did not significantly change IDO-2 and 5-lipoxygenase (5-LOX) expression, suggesting that proinflammatory parameters such as COX-2 and IDO-1 are specific for AMP-induced behaviors. Dextroamphetamine 181-184 prostaglandin-endoperoxide synthase 2 Mus musculus 148-153 31883280-4 2020 AMP-induced initial expression of COX-2 (15 min post-AMP) was earlier than that of IDO-1 (1 h post-AMP). Dextroamphetamine 0-3 prostaglandin-endoperoxide synthase 2 Mus musculus 34-39 31883280-4 2020 AMP-induced initial expression of COX-2 (15 min post-AMP) was earlier than that of IDO-1 (1 h post-AMP). Dextroamphetamine 0-3 indoleamine 2,3-dioxygenase 1 Mus musculus 83-88 31883280-4 2020 AMP-induced initial expression of COX-2 (15 min post-AMP) was earlier than that of IDO-1 (1 h post-AMP). Dextroamphetamine 53-56 prostaglandin-endoperoxide synthase 2 Mus musculus 34-39 31883280-4 2020 AMP-induced initial expression of COX-2 (15 min post-AMP) was earlier than that of IDO-1 (1 h post-AMP). Dextroamphetamine 53-56 prostaglandin-endoperoxide synthase 2 Mus musculus 34-39 31883280-5 2020 Mood stabilizer Li and COX-2 inhibitor meloxicam significantly attenuated COX-2 expression 15 min post-AMP, whereas IDO-1 inhibitor 1-methyl-DL-tryptophan (1-MT) did not affect COX-2 expression. Dextroamphetamine 103-106 prostaglandin-endoperoxide synthase 2 Mus musculus 23-28 31883280-5 2020 Mood stabilizer Li and COX-2 inhibitor meloxicam significantly attenuated COX-2 expression 15 min post-AMP, whereas IDO-1 inhibitor 1-methyl-DL-tryptophan (1-MT) did not affect COX-2 expression. Dextroamphetamine 103-106 prostaglandin-endoperoxide synthase 2 Mus musculus 74-79 31883280-5 2020 Mood stabilizer Li and COX-2 inhibitor meloxicam significantly attenuated COX-2 expression 15 min post-AMP, whereas IDO-1 inhibitor 1-methyl-DL-tryptophan (1-MT) did not affect COX-2 expression. Dextroamphetamine 103-106 prostaglandin-endoperoxide synthase 2 Mus musculus 74-79 31883280-6 2020 However, AMP-induced IDO-1 expression was significantly attenuated by Li, meloxocam or 1-MT, suggesting that COX-2 is an up-stream molecule for the induction of IDO-1 caused by AMP. Dextroamphetamine 9-12 indoleamine 2,3-dioxygenase 1 Mus musculus 21-26 31883280-6 2020 However, AMP-induced IDO-1 expression was significantly attenuated by Li, meloxocam or 1-MT, suggesting that COX-2 is an up-stream molecule for the induction of IDO-1 caused by AMP. Dextroamphetamine 9-12 prostaglandin-endoperoxide synthase 2 Mus musculus 109-114 31883280-6 2020 However, AMP-induced IDO-1 expression was significantly attenuated by Li, meloxocam or 1-MT, suggesting that COX-2 is an up-stream molecule for the induction of IDO-1 caused by AMP. Dextroamphetamine 9-12 indoleamine 2,3-dioxygenase 1 Mus musculus 161-166 31883280-6 2020 However, AMP-induced IDO-1 expression was significantly attenuated by Li, meloxocam or 1-MT, suggesting that COX-2 is an up-stream molecule for the induction of IDO-1 caused by AMP. Dextroamphetamine 177-180 indoleamine 2,3-dioxygenase 1 Mus musculus 21-26 31883280-6 2020 However, AMP-induced IDO-1 expression was significantly attenuated by Li, meloxocam or 1-MT, suggesting that COX-2 is an up-stream molecule for the induction of IDO-1 caused by AMP. Dextroamphetamine 177-180 prostaglandin-endoperoxide synthase 2 Mus musculus 109-114 31883280-6 2020 However, AMP-induced IDO-1 expression was significantly attenuated by Li, meloxocam or 1-MT, suggesting that COX-2 is an up-stream molecule for the induction of IDO-1 caused by AMP. Dextroamphetamine 177-180 indoleamine 2,3-dioxygenase 1 Mus musculus 161-166 31883280-7 2020 Consistently, co-immuoprecipitation between COX-2 and IDO-1 was observed at 30 min, 1 h, 3 h, and 6 h after the final AMP treatment. Dextroamphetamine 118-121 prostaglandin-endoperoxide synthase 2 Mus musculus 44-49 31883280-7 2020 Consistently, co-immuoprecipitation between COX-2 and IDO-1 was observed at 30 min, 1 h, 3 h, and 6 h after the final AMP treatment. Dextroamphetamine 118-121 indoleamine 2,3-dioxygenase 1 Mus musculus 54-59 31883280-10 2020 We, for the first time, report that mood stabilizer Li attenuates AMP-induced mania-like behavior via attenuation of interaction between COX-2 and IDO-1, and that the interaction of COX-2 and IDO-1 may be critical for the therapeutic intervention mediated by mood stabilizer. Dextroamphetamine 66-69 prostaglandin-endoperoxide synthase 2 Mus musculus 137-142 32163506-5 2020 After the motor activity test, the brains from d-amphetamine-treated animals were collected and processed for measurements of endocannabinoids and activation of Akt/GSK3beta, two molecular markers involved in the pathophysiology of schizophrenia. Dextroamphetamine 47-60 AKT serine/threonine kinase 1 Rattus norvegicus 161-164 32163506-5 2020 After the motor activity test, the brains from d-amphetamine-treated animals were collected and processed for measurements of endocannabinoids and activation of Akt/GSK3beta, two molecular markers involved in the pathophysiology of schizophrenia. Dextroamphetamine 47-60 glycogen synthase kinase 3 alpha Rattus norvegicus 165-173 31158395-7 2019 The administration of D-amph induced hyperactivity in the rats, as well increased the IL-4, IL-10, and TNF-alpha levels in the serum, frontal cortex, and striatum of rats compared to those of the controls, and treatment with Li plus Cel reversed these alterations. Dextroamphetamine 22-28 interleukin 4 Rattus norvegicus 86-90 31953370-13 2020 Last, intra-BLA infusions of d-amphetamine also intensified lever-pressing for the CS. Dextroamphetamine 29-42 citrate synthase Rattus norvegicus 83-85 31739003-2 2020 We examined the receptor-specific dopaminergic modulation of d-amphetamine (AMP)-altered DRL 10 sec (DRL-10 s) performance by locally infusing SCH23390 (SCH) and raclopride (RAC), DA D1 and D2 receptor antagonists, respectively, into the rat"s dSTR. Dextroamphetamine 61-74 defender against cell death 1 Rattus norvegicus 180-185 31739003-2 2020 We examined the receptor-specific dopaminergic modulation of d-amphetamine (AMP)-altered DRL 10 sec (DRL-10 s) performance by locally infusing SCH23390 (SCH) and raclopride (RAC), DA D1 and D2 receptor antagonists, respectively, into the rat"s dSTR. Dextroamphetamine 76-79 defender against cell death 1 Rattus norvegicus 180-185 31783118-8 2020 This is observed as D-amphetamine-induced hyperactivity, aberrant dopamine transporter function, and as increased D-amphetamine-induced dopamine release demonstrating that dopaminergic axon terminal function in the striatum of the Cdnf-/- mouse brain is altered. Dextroamphetamine 114-127 cerebral dopamine neurotrophic factor Mus musculus 231-235 31219577-6 2020 We also show that although the cerebral metabolic response to ketamine is preserved, the response to dextroamphetamine (d-amphetamine) is significantly attenuated in Map2k7+/- mice, supporting monoamine neurotransmitter system dysfunction but not glutamate/NMDA receptor (NMDA-R) dysfunction as a consequence of Map2k7 haploinsufficiency. Dextroamphetamine 101-118 mitogen-activated protein kinase kinase 7 Mus musculus 166-172 31219577-6 2020 We also show that although the cerebral metabolic response to ketamine is preserved, the response to dextroamphetamine (d-amphetamine) is significantly attenuated in Map2k7+/- mice, supporting monoamine neurotransmitter system dysfunction but not glutamate/NMDA receptor (NMDA-R) dysfunction as a consequence of Map2k7 haploinsufficiency. Dextroamphetamine 120-133 mitogen-activated protein kinase kinase 7 Mus musculus 166-172 31760073-0 2020 Indoleamine-2,3-dioxygenase-1 is a molecular target for the protective activity of mood stabilizers against mania-like behavior induced by d-amphetamine. Dextroamphetamine 139-152 indoleamine 2,3-dioxygenase 1 Mus musculus 0-29 31760073-2 2020 In the present study, we investigated whether a proinflammatory oxidative gene indoleamine-2,3-dioxygenase (IDO) is involved in AMPH-induced mitochondrial burden, and whether mood stabilizers (i.e., lithium and valproate) modulate IDO to protect against AMPH-induced mania-like behaviors. Dextroamphetamine 128-132 indoleamine 2,3-dioxygenase 1 Mus musculus 79-106 31760073-2 2020 In the present study, we investigated whether a proinflammatory oxidative gene indoleamine-2,3-dioxygenase (IDO) is involved in AMPH-induced mitochondrial burden, and whether mood stabilizers (i.e., lithium and valproate) modulate IDO to protect against AMPH-induced mania-like behaviors. Dextroamphetamine 128-132 indoleamine 2,3-dioxygenase 1 Mus musculus 108-111 31760073-3 2020 AMPH-induced IDO-1 expression was significantly greater than IDO-2 expression in the prefrontal cortex of wild type mice. Dextroamphetamine 0-4 indoleamine 2,3-dioxygenase 1 Mus musculus 13-18 31760073-9 2020 Collectively, our results suggest that mood stabilizers attenuate AMPH-induced mania-like behaviors via attenuation of IDO-1-dependent mitochondrial stress, highlighting IDO-1 as a novel molecular target for the protective potential of mood stabilizers. Dextroamphetamine 66-70 indoleamine 2,3-dioxygenase 1 Mus musculus 119-124 31760073-9 2020 Collectively, our results suggest that mood stabilizers attenuate AMPH-induced mania-like behaviors via attenuation of IDO-1-dependent mitochondrial stress, highlighting IDO-1 as a novel molecular target for the protective potential of mood stabilizers. Dextroamphetamine 66-70 indoleamine 2,3-dioxygenase 1 Mus musculus 170-175 33132261-4 2020 Pharmacologically, d-amphetamine competitively inhibits the dopamine transporter (DAT) and the noradrenaline transporter (NAT) to increase dopamine (DA) and noradrenaline (NA) concentrations in the synaptic cleft. Dextroamphetamine 19-32 solute carrier family 6 member 3 Homo sapiens 60-80 33132261-4 2020 Pharmacologically, d-amphetamine competitively inhibits the dopamine transporter (DAT) and the noradrenaline transporter (NAT) to increase dopamine (DA) and noradrenaline (NA) concentrations in the synaptic cleft. Dextroamphetamine 19-32 solute carrier family 6 member 3 Homo sapiens 82-85 33132261-4 2020 Pharmacologically, d-amphetamine competitively inhibits the dopamine transporter (DAT) and the noradrenaline transporter (NAT) to increase dopamine (DA) and noradrenaline (NA) concentrations in the synaptic cleft. Dextroamphetamine 19-32 solute carrier family 6 member 2 Homo sapiens 95-120 31158395-7 2019 The administration of D-amph induced hyperactivity in the rats, as well increased the IL-4, IL-10, and TNF-alpha levels in the serum, frontal cortex, and striatum of rats compared to those of the controls, and treatment with Li plus Cel reversed these alterations. Dextroamphetamine 22-28 interleukin 10 Rattus norvegicus 92-97 31158395-7 2019 The administration of D-amph induced hyperactivity in the rats, as well increased the IL-4, IL-10, and TNF-alpha levels in the serum, frontal cortex, and striatum of rats compared to those of the controls, and treatment with Li plus Cel reversed these alterations. Dextroamphetamine 22-28 tumor necrosis factor Rattus norvegicus 103-112 30170608-14 2018 RESULTS: ITGB4-/- mice exhibited mania-like behavior, including hyperlocomotion, D-amphetamine-induced hyperactivity, and reduced anxiety-like behavior. Dextroamphetamine 81-94 integrin beta 4 Mus musculus 9-14 30359640-7 2019 Finally, we have shown that, in alpha7 nAChR full knockout mice, the behavioral response to D-AMPH is attenuated, providing direct evidence for the role of alpha7 nAChRs on the physiological response to D-AMPH. Dextroamphetamine 92-98 cholinergic receptor, nicotinic, alpha polypeptide 7 Mus musculus 32-44 30359640-7 2019 Finally, we have shown that, in alpha7 nAChR full knockout mice, the behavioral response to D-AMPH is attenuated, providing direct evidence for the role of alpha7 nAChRs on the physiological response to D-AMPH. Dextroamphetamine 203-209 cholinergic receptor, nicotinic, alpha polypeptide 7 Mus musculus 32-44 31193305-10 2019 The treatment with VPA in the animals submitted to the administration of d-AMPH decreased the levels of ERK1, JNK-1, and JNK-2 phosphorylated in the hippocampus of the animals. Dextroamphetamine 73-79 mitogen activated protein kinase 3 Rattus norvegicus 104-108 31193305-10 2019 The treatment with VPA in the animals submitted to the administration of d-AMPH decreased the levels of ERK1, JNK-1, and JNK-2 phosphorylated in the hippocampus of the animals. Dextroamphetamine 73-79 mitogen-activated protein kinase 8 Rattus norvegicus 110-113 31193305-10 2019 The treatment with VPA in the animals submitted to the administration of d-AMPH decreased the levels of ERK1, JNK-1, and JNK-2 phosphorylated in the hippocampus of the animals. Dextroamphetamine 73-79 mitogen-activated protein kinase 8 Rattus norvegicus 121-124 31193305-11 2019 The treatment with Li decreased the JNK-1 phosphorylated in the hippocampus of the animals submitted to the animal model of mania induced by d-AMPH. Dextroamphetamine 141-147 mitogen-activated protein kinase 8 Rattus norvegicus 36-39 30733244-1 2019 Acute central nervous system exposure to dextroamphetamine (d-amphetamine) elicits a multitude of effects, including dual action on the dopamine transporter (DAT) to increase extracellular dopamine, and induction of a negative feedback response to limit the dopamine increase. Dextroamphetamine 41-58 solute carrier family 6 member 3 Rattus norvegicus 136-156 30733244-1 2019 Acute central nervous system exposure to dextroamphetamine (d-amphetamine) elicits a multitude of effects, including dual action on the dopamine transporter (DAT) to increase extracellular dopamine, and induction of a negative feedback response to limit the dopamine increase. Dextroamphetamine 41-58 solute carrier family 6 member 3 Rattus norvegicus 158-161 30733244-1 2019 Acute central nervous system exposure to dextroamphetamine (d-amphetamine) elicits a multitude of effects, including dual action on the dopamine transporter (DAT) to increase extracellular dopamine, and induction of a negative feedback response to limit the dopamine increase. Dextroamphetamine 60-73 solute carrier family 6 member 3 Rattus norvegicus 136-156 30733244-1 2019 Acute central nervous system exposure to dextroamphetamine (d-amphetamine) elicits a multitude of effects, including dual action on the dopamine transporter (DAT) to increase extracellular dopamine, and induction of a negative feedback response to limit the dopamine increase. Dextroamphetamine 60-73 solute carrier family 6 member 3 Rattus norvegicus 158-161 30733244-5 2019 In both rats and nonhuman primates (NHPs), d-amphetamine stimulation of dopamine outflow (reverse transport) through DAT was primarily responsible for the dose-linear increase in dopamine. Dextroamphetamine 43-56 solute carrier family 6 member 3 Rattus norvegicus 117-120 29195684-6 2018 IL-18KO mice showed impaired baseline PPI response, which was attenuated by d-amphetamine at a dose that did not modify PPI response in wild-type (WT) mice, suggesting a hypodopaminergic prefrontal cortex function in those mice. Dextroamphetamine 76-89 interleukin 18 Mus musculus 0-5 29511334-9 2018 D-amphetamine increased the level of Glu (p = .0001), Glx (p = .003), and tCr (p = .0067) in the dACC. Dextroamphetamine 0-13 T cell receptor beta variable 20/OR9-2 (non-functional) Homo sapiens 74-77 29511334-9 2018 D-amphetamine increased the level of Glu (p = .0001), Glx (p = .003), and tCr (p = .0067) in the dACC. Dextroamphetamine 0-13 Acetyl-CoA carboxylase Drosophila melanogaster 97-101 29511334-11 2018 Drug effects on Glu, tCr, and Glx were positively correlated with subjective drug responses, predicting both the duration of AMP liking (Glu: r = +.49, p = .02; tCr: r = +.41, p = .047) and the magnitude of peak drug high to MA (Glu: r = +.52, p = .016; Glx: r = +.42, p = .049). Dextroamphetamine 125-128 T cell receptor beta variable 20/OR9-2 (non-functional) Homo sapiens 21-24 29511334-11 2018 Drug effects on Glu, tCr, and Glx were positively correlated with subjective drug responses, predicting both the duration of AMP liking (Glu: r = +.49, p = .02; tCr: r = +.41, p = .047) and the magnitude of peak drug high to MA (Glu: r = +.52, p = .016; Glx: r = +.42, p = .049). Dextroamphetamine 125-128 T cell receptor beta variable 20/OR9-2 (non-functional) Homo sapiens 161-164 29511334-13 2018 We conclude that d-amphetamine increased the concentration of glutamate, Glx, and tCr in the dACC in male and female volunteers 21/2 hours after drug consumption. Dextroamphetamine 17-30 T cell receptor beta variable 20/OR9-2 (non-functional) Homo sapiens 82-85 29511334-13 2018 We conclude that d-amphetamine increased the concentration of glutamate, Glx, and tCr in the dACC in male and female volunteers 21/2 hours after drug consumption. Dextroamphetamine 17-30 Acetyl-CoA carboxylase Drosophila melanogaster 93-97 29355511-0 2018 Low-Dose d-Amphetamine Induced Regression of Liver Fat Deposits in Dercum Disease. Dextroamphetamine 9-22 FAT atypical cadherin 1 Homo sapiens 51-54 29355511-5 2018 Initiation of 10-20 mg d-amphetamine decreased liver lipid deposition from 16% to 4% in Case 1 and resolved fat deposits in Case 2 after ~1 year. Dextroamphetamine 23-36 FAT atypical cadherin 1 Homo sapiens 108-111 29355511-7 2018 Reduction of liver fat by d-amphetamine suggests a potential therapeutic role in nonalcoholic fatty liver disease. Dextroamphetamine 26-39 FAT atypical cadherin 1 Homo sapiens 19-22 29273699-0 2018 False-Positive Findings on Dopamine Transporter SPECT Due to Therapeutic Dextroamphetamine and Amphetamine. Dextroamphetamine 73-90 solute carrier family 6 member 3 Homo sapiens 27-47 29273699-3 2018 A patient had a false-positive dopamine transporter SPECT result while she was taking dextroamphetamine and amphetamine for attention-deficit hyperactivity disorder. Dextroamphetamine 86-103 solute carrier family 6 member 3 Homo sapiens 31-51 29170000-7 2018 This severe impact of a tonically impaired PPT cholinergic innervation was evidenced as the cholinergic interneuronal loss of the caudate putamen and as a suppressed c-Fos expression after stimulation by d-AMPH. Dextroamphetamine 204-210 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-171 29258368-7 2018 D-amphetamine is then further metabolized by CYP2D6. Dextroamphetamine 0-13 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 45-51 28452189-2 2017 This is important in light of the high prevalence of dexamphetamine (dAMPH) prescription for attention-deficit/hyperactivity disorder (ADHD), and its rising use as a recreational drug. Dextroamphetamine 53-67 Amphiphysin Drosophila melanogaster 69-74 29040826-3 2017 Previous studies indicate a role for endogenous opioid release and subsequent opioid receptor activation in some amphetamine effects; therefore, the current study examined the role of mu-opioid receptor activation in d-amphetamine treatment effects in an assay of cocaine-vs-food choice. Dextroamphetamine 217-230 opioid receptor mu 1 Macaca mulatta 184-202 28187857-8 2017 After a D-amphetamine challenge (5 mg/kg, intraperitoneal), Kmo-/- mice showed potentiated horizontal activity in the open field paradigm. Dextroamphetamine 8-21 kynurenine 3-monooxygenase (kynurenine 3-hydroxylase) Mus musculus 60-63 28860974-11 2017 Moreover, we demonstrated a pathway-specific activation pattern of D1 MSNs and D2 MSNs in a manic like mouse model induced by D-Amphetamine by utilizing this double transgenic mice and c-fos immunoreactivity. Dextroamphetamine 126-139 FBJ osteosarcoma oncogene Mus musculus 185-190 26946431-11 2016 Together, these findings suggest that mGluR5 activation in the NAc is not essential for the expression of mating, but that experience-induced reduction in mGluR5 protein may contribute to the cross-sensitization of amph responses by sexual experience and abstinence. Dextroamphetamine 215-219 glutamate receptor, ionotropic, kainate 1 Mus musculus 155-161 29564380-6 2017 5-day D-amphetamine treatment (5 mg/kg, s.c.) increased striatal PDE10A BPND compared to the baseline (+24 %, p = 0.03). Dextroamphetamine 6-19 phosphodiesterase 10A Rattus norvegicus 65-71 29564380-7 2017 Treatment with the selective D2 antagonist SCH23390 (1 mg/kg) and D-amphetamine decreased PDE10A binding (-22 %, p = 0.03). Dextroamphetamine 66-79 phosphodiesterase 10A Homo sapiens 90-96 29564380-10 2017 Conclusions: Repeated D-amphetamine treatment significantly increased PDE10A binding, which is not observed upon selective D1 receptor blocking. Dextroamphetamine 22-35 phosphodiesterase 10A Homo sapiens 70-76 27791160-1 2016 Dopamine (DA) promotes wakefulness, and DA transporter inhibitors such as dextroamphetamine and methylphenidate are effective for increasing arousal and inducing reanimation, or active emergence from general anesthesia. Dextroamphetamine 74-91 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 Mus musculus 40-54 27822508-7 2016 In locomotor studies, CNO alone (at 1, 2, and 5 mg/kg) had no effect on spontaneous locomotion, but 5 mg/kg CNO pretreatment significantly attenuated d-amphetamine-induced hyperlocomotion. Dextroamphetamine 150-163 biogenesis of lysosomal organelles complex 1 subunit 4 Rattus norvegicus 108-111 27822508-8 2016 In line with the behavioral results, fast-scan cyclic voltammetry found that 5 mg/kg CNO significantly attenuated the d-amphetamine-induced increase in evoked dopamine. Dextroamphetamine 118-131 biogenesis of lysosomal organelles complex 1 subunit 4 Rattus norvegicus 85-88 29363908-6 2017 Analysis of the pH3S10 and c-Fos expression levels in the group of D-amphetamine administered rats provided evidence of enhanced expression of these proteins in the regions of neurogenesis occurrence in rats. Dextroamphetamine 67-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 27-32 27771748-9 2017 CONCLUSIONS: In concert with a previous GWAS result, this candidate gene study provides convergent evidence implicating CDH13 rs3784943 variant in d-amphetamine"s drug effect profile and suggests generalization to Asian populations. Dextroamphetamine 147-160 cadherin 13 Homo sapiens 120-125 26946431-9 2016 Instead, sexually naive animals that received NAc mGluR5 antagonists without mating demonstrated sensitized amph-induced locomotor responses and enhanced CPP on par with sexually experienced males. Dextroamphetamine 108-112 glutamate receptor, ionotropic, kainate 1 Mus musculus 50-56 26453694-6 2016 Qualitative analysis of exploration revealed tolerance to D-amphetamine effects on entropy in methamphetamine-treated brain-derived neurotrophic factor heterozygous mice, but not wild-type mice. Dextroamphetamine 58-71 brain derived neurotrophic factor Mus musculus 118-151 26136480-10 2015 d-Amphetamine (potent sympathomimetic) caused hyperthermia in WT mice, which was reduced in TRPV1 KO mice, suggesting a decreased sympathetic drive in KOs. Dextroamphetamine 0-13 transient receptor potential cation channel, subfamily V, member 1 Mus musculus 92-97 25502294-0 2016 Role of P2X7 Receptor in an Animal Model of Mania Induced by D-Amphetamine. Dextroamphetamine 61-74 purinergic receptor P2X, ligand-gated ion channel, 7 Mus musculus 8-21 25502294-4 2016 Likewise, P2X7R participated in the AMPH-induced increase of the proinflammatory and excitotoxic environment, as demonstrated by the reversal of IL-1beta, TNF-alpha, and TBARS levels caused by P2X7R blocking. Dextroamphetamine 36-40 purinergic receptor P2X, ligand-gated ion channel, 7 Mus musculus 10-15 25502294-4 2016 Likewise, P2X7R participated in the AMPH-induced increase of the proinflammatory and excitotoxic environment, as demonstrated by the reversal of IL-1beta, TNF-alpha, and TBARS levels caused by P2X7R blocking. Dextroamphetamine 36-40 interleukin 1 beta Mus musculus 145-153 25502294-4 2016 Likewise, P2X7R participated in the AMPH-induced increase of the proinflammatory and excitotoxic environment, as demonstrated by the reversal of IL-1beta, TNF-alpha, and TBARS levels caused by P2X7R blocking. Dextroamphetamine 36-40 tumor necrosis factor Mus musculus 155-164 25502294-4 2016 Likewise, P2X7R participated in the AMPH-induced increase of the proinflammatory and excitotoxic environment, as demonstrated by the reversal of IL-1beta, TNF-alpha, and TBARS levels caused by P2X7R blocking. Dextroamphetamine 36-40 purinergic receptor P2X, ligand-gated ion channel, 7 Mus musculus 193-198 25502294-5 2016 Our results support the hypothesis that P2X7R plays a role in the neuroinflammation induced by AMPH in a preclinical model of mania, which could explain the altered behavior. Dextroamphetamine 95-99 purinergic receptor P2X, ligand-gated ion channel, 7 Mus musculus 40-45 26640076-2 2016 Accordingly, the present study aimed to investigate the role of TAAR1 in the effects of psychostimulants by analyzing context-dependent sensitization and conditioned place preference (CPP) to d-amphetamine (AMPH) in TAAR1-KO mice. Dextroamphetamine 192-205 trace amine-associated receptor 1 Mus musculus 216-221 26640076-2 2016 Accordingly, the present study aimed to investigate the role of TAAR1 in the effects of psychostimulants by analyzing context-dependent sensitization and conditioned place preference (CPP) to d-amphetamine (AMPH) in TAAR1-KO mice. Dextroamphetamine 207-211 trace amine-associated receptor 1 Mus musculus 216-221 26640076-4 2016 In the CPP test, TAAR1-KO animals were also more sensitive to priming-induced reinstatement of AMPH-induced conditioned place preference (CPP) than wild type mice. Dextroamphetamine 95-99 trace amine-associated receptor 1 Mus musculus 17-22 26640076-5 2016 Importantly, saline-treated and AMPH-treated mice lacking TAAR1 demonstrated significant alterations in the total levels and phosphorylation of the critical subunit of NMDA glutamate receptors, GluN1, in the striatum, suggesting a role of TAAR1 in the modulation of frontostriatal glutamate transmission; this effect could underlie the observed alterations in conditioning processes. Dextroamphetamine 32-36 trace amine-associated receptor 1 Mus musculus 58-63 26640076-5 2016 Importantly, saline-treated and AMPH-treated mice lacking TAAR1 demonstrated significant alterations in the total levels and phosphorylation of the critical subunit of NMDA glutamate receptors, GluN1, in the striatum, suggesting a role of TAAR1 in the modulation of frontostriatal glutamate transmission; this effect could underlie the observed alterations in conditioning processes. Dextroamphetamine 32-36 glutamate receptor, ionotropic, NMDA1 (zeta 1) Mus musculus 194-199 26640076-5 2016 Importantly, saline-treated and AMPH-treated mice lacking TAAR1 demonstrated significant alterations in the total levels and phosphorylation of the critical subunit of NMDA glutamate receptors, GluN1, in the striatum, suggesting a role of TAAR1 in the modulation of frontostriatal glutamate transmission; this effect could underlie the observed alterations in conditioning processes. Dextroamphetamine 32-36 trace amine-associated receptor 1 Mus musculus 239-244 26640076-6 2016 In conclusion, our data suggest that TAAR1 receptors play an inhibitory role with respect to conditioned responses to AMPH by modulating, at least in part, corticostriatal glutamate transmission. Dextroamphetamine 118-122 trace amine-associated receptor 1 Mus musculus 37-42 26092248-4 2015 In the present study, we show that heterozygous CTSD-deficient (CTSD HET) mice display an overall behavioral profile that is similar to human mania, including hyperlocomotion, d-amphetamine-induced hyperactivity, sleep-disturbance, and reduced anxiety-like behavior. Dextroamphetamine 176-189 cathepsin D Mus musculus 48-52 26577247-8 2016 Beta 1 and beta 2 power were reduced by dexamphetamine at Frontal ICs, while beta 2 and gamma power was enhanced by dexamphetamine in posterior regions, including the parietal, occipital-temporal, and occipital regions. Dextroamphetamine 40-54 potassium calcium-activated channel subfamily M regulatory beta subunit 1 Homo sapiens 0-6 26577247-8 2016 Beta 1 and beta 2 power were reduced by dexamphetamine at Frontal ICs, while beta 2 and gamma power was enhanced by dexamphetamine in posterior regions, including the parietal, occipital-temporal, and occipital regions. Dextroamphetamine 40-54 potassium calcium-activated channel subfamily M regulatory beta subunit 2 Homo sapiens 11-17 26577247-8 2016 Beta 1 and beta 2 power were reduced by dexamphetamine at Frontal ICs, while beta 2 and gamma power was enhanced by dexamphetamine in posterior regions, including the parietal, occipital-temporal, and occipital regions. Dextroamphetamine 116-130 potassium calcium-activated channel subfamily M regulatory beta subunit 2 Homo sapiens 77-83 25563230-1 2015 D-amphetamine (dAMPH) and methylphenidate (MPH) are stimulants used in the treatment of Attention Deficit Hyperactivity Disorder (ADHD). Dextroamphetamine 0-13 Amphiphysin Drosophila melanogaster 15-20 25986696-0 2015 D-Amphetamine withdrawal-induced decreases in brain-derived neurotrophic factor in sprague-dawley rats are reversed by treatment with ketamine. Dextroamphetamine 0-13 brain-derived neurotrophic factor Rattus norvegicus 46-79 25986696-4 2015 The goals of this study were to examine BDNF levels throughout the limbic system following D-AMPH withdrawal and determine whether ketamine treatment would alter D-AMPH-induced changes in BDNF. Dextroamphetamine 162-168 brain-derived neurotrophic factor Rattus norvegicus 188-192 25986696-6 2015 Our data show that at 24 h post-D-AMPH, BDNF levels were increased in the nucleus accumbens and decreased in the hippocampus. Dextroamphetamine 32-38 brain-derived neurotrophic factor Rattus norvegicus 40-44 25986696-7 2015 At 4 d post-D-AMPH, BDNF protein levels were decreased in all areas examined, and these decreases were reversed by treatment with ketamine. Dextroamphetamine 12-18 brain-derived neurotrophic factor Rattus norvegicus 20-24 25986696-8 2015 These data suggest that diminished BDNF may contribute to the negative affect seen following D-AMPH withdrawal, and that ketamine treatment could offer relief from these symptoms. Dextroamphetamine 93-99 brain-derived neurotrophic factor Rattus norvegicus 35-39 25970245-2 2015 Here we determine X-ray crystal structures of the Drosophila melanogaster dopamine transporter (dDAT) bound to its substrate dopamine, a substrate analogue 3,4-dichlorophenethylamine, the psychostimulants d-amphetamine and methamphetamine, or to cocaine and cocaine analogues. Dextroamphetamine 205-218 Dopamine transporter Drosophila melanogaster 74-94 25999268-4 2015 Additionally, a protective effect was observed following repeated intranasal administration in 6-OHDA lesioned rats, as suggested by: a significant decrease in d-amphetamine-induced rotation at 2 weeks; a decrease in DA turnover in the lesioned striatum; and an increased sparing of tyrosine hydroxylase (TH) positive (+) neurons in a specific sub-region of the lesioned substantia nigra pars compacta (SNpc). Dextroamphetamine 160-173 tyrosine hydroxylase Rattus norvegicus 283-303 25999268-4 2015 Additionally, a protective effect was observed following repeated intranasal administration in 6-OHDA lesioned rats, as suggested by: a significant decrease in d-amphetamine-induced rotation at 2 weeks; a decrease in DA turnover in the lesioned striatum; and an increased sparing of tyrosine hydroxylase (TH) positive (+) neurons in a specific sub-region of the lesioned substantia nigra pars compacta (SNpc). Dextroamphetamine 160-173 tyrosine hydroxylase Rattus norvegicus 305-307 25959066-3 2015 CD-1 mice were subcutaneously dosed for 5 days with 1.8 mg/kg d-amphetamine or vehicle. Dextroamphetamine 62-75 CD1 antigen complex Mus musculus 0-4 25970245-2 2015 Here we determine X-ray crystal structures of the Drosophila melanogaster dopamine transporter (dDAT) bound to its substrate dopamine, a substrate analogue 3,4-dichlorophenethylamine, the psychostimulants d-amphetamine and methamphetamine, or to cocaine and cocaine analogues. Dextroamphetamine 205-218 Dopamine transporter Drosophila melanogaster 96-100 25489246-8 2014 The demonstration that aminopeptidase-like activity in red blood cell cytosol is responsible for the hydrolysis of LDX extends our understanding of the smooth and consistent systemic delivery of d-amphetamine by LDX and the long daily duration of efficacy of the drug in relieving the symptoms of attention-deficit/hyperactivity disorder. Dextroamphetamine 195-208 carboxypeptidase Q Homo sapiens 23-37 25762659-3 2015 Here, we demonstrate that the psychostimulant d-amphetamine (d-amph) markedly increases rpS6 phosphorylation at Ser235/236 sites in both crude and synaptoneurosomal preparations of the mouse striatum. Dextroamphetamine 46-59 ribosomal protein S6 Mus musculus 88-92 25762659-3 2015 Here, we demonstrate that the psychostimulant d-amphetamine (d-amph) markedly increases rpS6 phosphorylation at Ser235/236 sites in both crude and synaptoneurosomal preparations of the mouse striatum. Dextroamphetamine 46-52 ribosomal protein S6 Mus musculus 88-92 25762659-5 2015 By developing a novel assay to label nascent peptidic chains, we show that the rpS6 phosphorylation induced in striatonigral MSNs by d-amph, as well as in striatopallidal MSNs by the antipsychotic haloperidol or in both subtypes by papaverine, is not correlated with the translation of global or 5" terminal oligopyrimidine tract mRNAs. Dextroamphetamine 133-139 ribosomal protein S6 Mus musculus 79-83 24672455-7 2014 Acute and sensitized hyperlocomotion induced by d-amphetamine and cocaine are dramatically reduced in T-type Cav3.2 deficient mice. Dextroamphetamine 48-61 calcium channel, voltage-dependent, T type, alpha 1H subunit Mus musculus 102-115 24997241-8 2014 In addition, hGDNF protected nigral DA neurons and striatal DA fibers, and attenuated d-amphetamine-induced rotational asymmetry in the 6-OHDA lesioned rats. Dextroamphetamine 86-99 glial cell derived neurotrophic factor Homo sapiens 13-18 25246058-6 2014 Intrathecal d-amphetamine (AMPH, 30 muM) did not but higher AMPH concentration (100 muM) induced SRP in TS-induced reflex activity. Dextroamphetamine 60-64 latexin Homo sapiens 84-87 25246058-7 2014 H89 (10 muM, a protein kinase A inhibitor), but not chelerythrine chloride (CTC, 10 muM, a protein kinase C inhibitor), prevented the 100 muM AMPH-elicited SRP. Dextroamphetamine 142-146 latexin Homo sapiens 8-11 25246058-9 2014 The co-administration of 10 muM forskolin and 30 muM AMPH induced SRP in TS-induced reflex activity. Dextroamphetamine 53-57 latexin Homo sapiens 49-52 24535653-8 2014 RESULTS: FR increased GluA1 in the PSD, and D-amphetamine increased p-Ser845-GluA1, GluA1, GluA2, but not GluA3, with a greater effect in FR than AL rats. Dextroamphetamine 44-57 glutamate ionotropic receptor AMPA type subunit 1 Rattus norvegicus 77-82 24535653-8 2014 RESULTS: FR increased GluA1 in the PSD, and D-amphetamine increased p-Ser845-GluA1, GluA1, GluA2, but not GluA3, with a greater effect in FR than AL rats. Dextroamphetamine 44-57 glutamate ionotropic receptor AMPA type subunit 1 Rattus norvegicus 77-82 24535653-8 2014 RESULTS: FR increased GluA1 in the PSD, and D-amphetamine increased p-Ser845-GluA1, GluA1, GluA2, but not GluA3, with a greater effect in FR than AL rats. Dextroamphetamine 44-57 glutamate ionotropic receptor AMPA type subunit 2 Rattus norvegicus 91-96 24535653-8 2014 RESULTS: FR increased GluA1 in the PSD, and D-amphetamine increased p-Ser845-GluA1, GluA1, GluA2, but not GluA3, with a greater effect in FR than AL rats. Dextroamphetamine 44-57 glutamate ionotropic receptor AMPA type subunit 3 Rattus norvegicus 106-111 24535653-11 2014 The D-amphetamine-induced increase in synaptic p-Ser845-GluA1, GluA1, and GluA2 may contribute to the rewarding effect of D-amphetamine, but may also be a mechanism of synaptic strengthening and behavior modification. Dextroamphetamine 4-17 glutamate ionotropic receptor AMPA type subunit 1 Rattus norvegicus 56-61 24535653-11 2014 The D-amphetamine-induced increase in synaptic p-Ser845-GluA1, GluA1, and GluA2 may contribute to the rewarding effect of D-amphetamine, but may also be a mechanism of synaptic strengthening and behavior modification. Dextroamphetamine 4-17 glutamate ionotropic receptor AMPA type subunit 1 Rattus norvegicus 63-68 24535653-11 2014 The D-amphetamine-induced increase in synaptic p-Ser845-GluA1, GluA1, and GluA2 may contribute to the rewarding effect of D-amphetamine, but may also be a mechanism of synaptic strengthening and behavior modification. Dextroamphetamine 4-17 glutamate ionotropic receptor AMPA type subunit 2 Rattus norvegicus 74-79 24535653-11 2014 The D-amphetamine-induced increase in synaptic p-Ser845-GluA1, GluA1, and GluA2 may contribute to the rewarding effect of D-amphetamine, but may also be a mechanism of synaptic strengthening and behavior modification. Dextroamphetamine 122-135 glutamate ionotropic receptor AMPA type subunit 1 Rattus norvegicus 56-61 24535653-11 2014 The D-amphetamine-induced increase in synaptic p-Ser845-GluA1, GluA1, and GluA2 may contribute to the rewarding effect of D-amphetamine, but may also be a mechanism of synaptic strengthening and behavior modification. Dextroamphetamine 122-135 glutamate ionotropic receptor AMPA type subunit 1 Rattus norvegicus 63-68 24535653-11 2014 The D-amphetamine-induced increase in synaptic p-Ser845-GluA1, GluA1, and GluA2 may contribute to the rewarding effect of D-amphetamine, but may also be a mechanism of synaptic strengthening and behavior modification. Dextroamphetamine 122-135 glutamate ionotropic receptor AMPA type subunit 2 Rattus norvegicus 74-79 24440750-10 2014 These findings support the notion that BDNF is involved in drug-seeking behavior and indicate that d-AMP reinstatement after extinction may be linked to an increase in BDNF mRNA expression in the mPFC. Dextroamphetamine 99-104 brain-derived neurotrophic factor Rattus norvegicus 168-172 24287377-0 2014 Stress and withdrawal from d-amphetamine alter 5-HT2A receptor mRNA expression in the prefrontal cortex. Dextroamphetamine 27-40 5-hydroxytryptamine receptor 2A Homo sapiens 47-62 24287377-3 2014 5-HT2A receptor (5-HT2AR) mRNA expression in the prefrontal cortex (PFC) is diminished following withdrawal from d-amphetamine (AMPH) and may underlie the emotional and cognitive impairments observed in psychostimulant withdrawal, but whether stress affects 5-HT2AR mRNA expression during psychostimulant withdrawal is unknown. Dextroamphetamine 113-126 5-hydroxytryptamine receptor 2A Homo sapiens 0-15 24287377-3 2014 5-HT2A receptor (5-HT2AR) mRNA expression in the prefrontal cortex (PFC) is diminished following withdrawal from d-amphetamine (AMPH) and may underlie the emotional and cognitive impairments observed in psychostimulant withdrawal, but whether stress affects 5-HT2AR mRNA expression during psychostimulant withdrawal is unknown. Dextroamphetamine 113-126 5-hydroxytryptamine receptor 2A Homo sapiens 17-24 24287377-3 2014 5-HT2A receptor (5-HT2AR) mRNA expression in the prefrontal cortex (PFC) is diminished following withdrawal from d-amphetamine (AMPH) and may underlie the emotional and cognitive impairments observed in psychostimulant withdrawal, but whether stress affects 5-HT2AR mRNA expression during psychostimulant withdrawal is unknown. Dextroamphetamine 113-126 5-hydroxytryptamine receptor 2A Homo sapiens 258-265 24287377-3 2014 5-HT2A receptor (5-HT2AR) mRNA expression in the prefrontal cortex (PFC) is diminished following withdrawal from d-amphetamine (AMPH) and may underlie the emotional and cognitive impairments observed in psychostimulant withdrawal, but whether stress affects 5-HT2AR mRNA expression during psychostimulant withdrawal is unknown. Dextroamphetamine 128-132 5-hydroxytryptamine receptor 2A Homo sapiens 0-15 24287377-3 2014 5-HT2A receptor (5-HT2AR) mRNA expression in the prefrontal cortex (PFC) is diminished following withdrawal from d-amphetamine (AMPH) and may underlie the emotional and cognitive impairments observed in psychostimulant withdrawal, but whether stress affects 5-HT2AR mRNA expression during psychostimulant withdrawal is unknown. Dextroamphetamine 128-132 5-hydroxytryptamine receptor 2A Homo sapiens 17-24 24287377-3 2014 5-HT2A receptor (5-HT2AR) mRNA expression in the prefrontal cortex (PFC) is diminished following withdrawal from d-amphetamine (AMPH) and may underlie the emotional and cognitive impairments observed in psychostimulant withdrawal, but whether stress affects 5-HT2AR mRNA expression during psychostimulant withdrawal is unknown. Dextroamphetamine 128-132 5-hydroxytryptamine receptor 2A Homo sapiens 258-265 24440750-0 2014 Elevated BDNF mRNA expression in the medial prefrontal cortex after d-amphetamine reinstated conditioned place preference in rats. Dextroamphetamine 68-81 brain-derived neurotrophic factor Rattus norvegicus 9-13 24440750-3 2014 To fill this gap, the present study was designed to test whether BDNF mRNA expression levels in the DA terminal regions were changed specifically by d-AMP-induced conditioned place preference (CPP) followed by drug-primed reinstatement. Dextroamphetamine 149-154 brain-derived neurotrophic factor Rattus norvegicus 65-69 24807738-4 2014 This paper discusses the changes in other markers of oxidative stress - the isozymes of superoxide dismutase Mn-SOD and Cu/Zn-SOD - in nicotine- and nicotine + D-amphetamine-treated rats. Dextroamphetamine 160-173 superoxide dismutase 2 Rattus norvegicus 109-115 24287377-4 2014 The goal of this study was to examine the impact of forced swim test (FST) exposure during AMPH withdrawal on 5-HT2AR mRNA expression in PFC. Dextroamphetamine 91-95 5-hydroxytryptamine receptor 2A Homo sapiens 110-117 24287377-8 2014 At 4 days of withdrawal, AMPH-treated animals showed greater 5-HT2AR mRNA expression relative to saline-treated animals in the PFC, an effect that was diminished by exposure to FST. Dextroamphetamine 25-29 5-hydroxytryptamine receptor 2A Homo sapiens 61-68 24287377-9 2014 These data indicate that stress and short-term AMPH withdrawal affect prefrontal 5-HT2AR mRNA expression to a similar degree, and stress experienced during long-term AMPH withdrawal can diminish the recovery of 5-HT2AR mRNA expression. Dextroamphetamine 47-51 5-hydroxytryptamine receptor 2A Homo sapiens 81-88 24287377-9 2014 These data indicate that stress and short-term AMPH withdrawal affect prefrontal 5-HT2AR mRNA expression to a similar degree, and stress experienced during long-term AMPH withdrawal can diminish the recovery of 5-HT2AR mRNA expression. Dextroamphetamine 166-170 5-hydroxytryptamine receptor 2A Homo sapiens 211-218 24287377-10 2014 Together, these data suggest that exposure to stress during extended AMPH withdrawal could prolong withdrawal-induced, 5-HT2AR mRNA expression which could be related to 5-HT2AR mediated deficits. Dextroamphetamine 69-73 5-hydroxytryptamine receptor 2A Homo sapiens 119-126 24287377-10 2014 Together, these data suggest that exposure to stress during extended AMPH withdrawal could prolong withdrawal-induced, 5-HT2AR mRNA expression which could be related to 5-HT2AR mediated deficits. Dextroamphetamine 69-73 5-hydroxytryptamine receptor 2A Homo sapiens 169-176 23422792-1 2013 Drugs that induce psychosis, such as D-amphetamine (AMP), and those that alleviate it, such as antipsychotics, are suggested to exert behavioral effects via dopamine receptor D2 (D2). Dextroamphetamine 37-50 dopamine receptor D2 Mus musculus 157-177 23245536-0 2013 Evaluation of acetylcholinesterase in an animal model of mania induced by D-amphetamine. Dextroamphetamine 74-87 acetylcholinesterase Rattus norvegicus 14-34 23245536-1 2013 The present study aims to investigate the effects of mood stabilizers, lithium (Li) and valproate (VPA), on acetylcholinesterase (AChE) activity in the brains of rats subjected to an animal model of mania induced by D-amphetamine (D-AMPH). Dextroamphetamine 216-229 acetylcholinesterase Rattus norvegicus 108-128 23245536-1 2013 The present study aims to investigate the effects of mood stabilizers, lithium (Li) and valproate (VPA), on acetylcholinesterase (AChE) activity in the brains of rats subjected to an animal model of mania induced by D-amphetamine (D-AMPH). Dextroamphetamine 231-237 acetylcholinesterase Rattus norvegicus 108-128 23652158-5 2013 Administration of d-AMPH (3 mg/kg, intraperitoneally) to Sprague-Dawley rats resulted in a concomitant decrease in levels of phosphorylated (p) Akt as well as p-FoxO1 in the striatum, whereas lithium chloride (LiCl,100 mg/kg, intraperitoneally) exerted the opposite effect, that is, it increased levels of p-Akt and p-FoxO1. Dextroamphetamine 18-24 AKT serine/threonine kinase 1 Rattus norvegicus 144-147 23652158-5 2013 Administration of d-AMPH (3 mg/kg, intraperitoneally) to Sprague-Dawley rats resulted in a concomitant decrease in levels of phosphorylated (p) Akt as well as p-FoxO1 in the striatum, whereas lithium chloride (LiCl,100 mg/kg, intraperitoneally) exerted the opposite effect, that is, it increased levels of p-Akt and p-FoxO1. Dextroamphetamine 18-24 forkhead box O1 Rattus norvegicus 161-166 23652158-5 2013 Administration of d-AMPH (3 mg/kg, intraperitoneally) to Sprague-Dawley rats resulted in a concomitant decrease in levels of phosphorylated (p) Akt as well as p-FoxO1 in the striatum, whereas lithium chloride (LiCl,100 mg/kg, intraperitoneally) exerted the opposite effect, that is, it increased levels of p-Akt and p-FoxO1. Dextroamphetamine 18-24 AKT serine/threonine kinase 1 Rattus norvegicus 308-311 23652158-5 2013 Administration of d-AMPH (3 mg/kg, intraperitoneally) to Sprague-Dawley rats resulted in a concomitant decrease in levels of phosphorylated (p) Akt as well as p-FoxO1 in the striatum, whereas lithium chloride (LiCl,100 mg/kg, intraperitoneally) exerted the opposite effect, that is, it increased levels of p-Akt and p-FoxO1. Dextroamphetamine 18-24 forkhead box O1 Rattus norvegicus 318-323 24099355-7 2013 Administration of 1 mg/kg d-amphetamine increased the mean wait time in both Ts65Dn and LC, though it was statistically significant only for the LC. Dextroamphetamine 26-39 reciprocal translocation, Chr 16, cytogenetic band C3-4; and Chr 17, cytogenetic band A2, Davisson 65 Mus musculus 77-83 23757186-6 2013 Amphetamine, dextroamphetamine, and methylphenidate act as substrates for the cellular monoamine transporter, especially the dopamine transporter (DAT) and less so the norepinephrine (NET) and serotonin transporter. Dextroamphetamine 13-30 solute carrier family 6 member 3 Homo sapiens 125-145 23757186-6 2013 Amphetamine, dextroamphetamine, and methylphenidate act as substrates for the cellular monoamine transporter, especially the dopamine transporter (DAT) and less so the norepinephrine (NET) and serotonin transporter. Dextroamphetamine 13-30 solute carrier family 6 member 3 Homo sapiens 147-150 23757186-6 2013 Amphetamine, dextroamphetamine, and methylphenidate act as substrates for the cellular monoamine transporter, especially the dopamine transporter (DAT) and less so the norepinephrine (NET) and serotonin transporter. Dextroamphetamine 13-30 solute carrier family 6 member 4 Homo sapiens 193-214 23422792-8 2013 AMP disruption of LI was attenuated in mice lacking dopamine receptor D1 (Drd1-/-), suggesting that D1 may play a role in AMP disruption of LI. Dextroamphetamine 0-3 dopamine receptor D1 Mus musculus 52-72 23422792-8 2013 AMP disruption of LI was attenuated in mice lacking dopamine receptor D1 (Drd1-/-), suggesting that D1 may play a role in AMP disruption of LI. Dextroamphetamine 0-3 dopamine receptor D1 Mus musculus 74-78 23422792-8 2013 AMP disruption of LI was attenuated in mice lacking dopamine receptor D1 (Drd1-/-), suggesting that D1 may play a role in AMP disruption of LI. Dextroamphetamine 122-125 dopamine receptor D1 Mus musculus 52-72 23422792-8 2013 AMP disruption of LI was attenuated in mice lacking dopamine receptor D1 (Drd1-/-), suggesting that D1 may play a role in AMP disruption of LI. Dextroamphetamine 122-125 dopamine receptor D1 Mus musculus 74-78 23422792-10 2013 Remarkably, both haloperidol and clozapine attenuated AMP disruption of LI in Drd2-/-. Dextroamphetamine 54-57 dopamine receptor D2 Mus musculus 78-82 23623962-5 2013 PCB-exposed males showed greater activation to the initial acute AMPH injection, but sensitization occurred later and was blunted relative to controls. Dextroamphetamine 65-69 pyruvate carboxylase Rattus norvegicus 0-3 23623962-8 2013 Overall, these results indicated developmental PCB exposure can alter the motor-stimulating effects of repeated AMPH injections. Dextroamphetamine 112-116 pyruvate carboxylase Rattus norvegicus 47-50 23422792-1 2013 Drugs that induce psychosis, such as D-amphetamine (AMP), and those that alleviate it, such as antipsychotics, are suggested to exert behavioral effects via dopamine receptor D2 (D2). Dextroamphetamine 52-55 dopamine receptor D2 Mus musculus 157-177 23422792-6 2013 AMP disruption of LI was seen in both wild-type (WT) and Drd2-/-. Dextroamphetamine 0-3 dopamine receptor D2 Mus musculus 57-61 23422792-7 2013 This was in contrast to AMP-induced locomotor hyperactivity, which was reduced in Drd2-/-. Dextroamphetamine 24-27 dopamine receptor D2 Mus musculus 82-86 23518151-5 2013 Chronic administration shifted the phase of Per1 and Per2 expressions from a nocturnal to diurnal pattern and advance shifted the peak of Rev-erbalpha in d-amphetamine-treated animals. Dextroamphetamine 154-167 nuclear receptor subfamily 1, group D, member 1 Rattus norvegicus 138-150 23142493-2 2013 Preclinical studies have shown that even relatively low doses of d-amphetamine (dAMPH) (equivalent to doses used in clinical Practice) can lead to DA neurotoxicity in rodents and non-human primates (Ricaurte et al., 2005). Dextroamphetamine 65-78 Amphiphysin Drosophila melanogaster 80-85 23219665-8 2013 Less 6-OHDA was required in tg(Prnp-SNCA A30P) Tg (1 mug) than in WT (3mug) mice for an ipsilateral rotational bias by d-amphetamine. Dextroamphetamine 119-132 prion protein Mus musculus 31-35 23423476-7 2013 In addition, cell-type specific extracellular signal-regulated kinase (ERK) phosphorylation in the NAc subterritories was analyzed following acute administration of SKF81297 (a D1R-like agonist), quinpirole (a D2 receptors (D2R)-like agonist), apomorphine (a non-selective DA receptor agonist), raclopride (a D2R-like antagonist), and psychostimulant drugs, including cocaine and d-amphetamine. Dextroamphetamine 380-393 mitogen-activated protein kinase 1 Mus musculus 71-74 23178911-10 2013 d-Amphetamine dose-dependently increased survival and differentiation of BrdU cells into neurons and increased number of DCX cells without affecting the number of Ki67 cells. Dextroamphetamine 0-13 doublecortin Mus musculus 121-124 23178911-11 2013 Low doses of d-amphetamine decreased c-Fos and DeltaFosB in the granule layer. Dextroamphetamine 13-26 FBJ osteosarcoma oncogene Mus musculus 37-42 23219665-8 2013 Less 6-OHDA was required in tg(Prnp-SNCA A30P) Tg (1 mug) than in WT (3mug) mice for an ipsilateral rotational bias by d-amphetamine. Dextroamphetamine 119-132 synuclein, alpha Mus musculus 36-40 23184940-5 2013 In addition, Tat-Sab(KIM1) decreased the d-amphetamine-induced unilateral rotations associated with the lesion by 30% (p < 0.05). Dextroamphetamine 41-54 SH3 domain binding protein 5 Homo sapiens 17-20 23184940-5 2013 In addition, Tat-Sab(KIM1) decreased the d-amphetamine-induced unilateral rotations associated with the lesion by 30% (p < 0.05). Dextroamphetamine 41-54 hepatitis A virus cellular receptor 1 Homo sapiens 21-25 23022501-4 2012 The goal of the present study was thus to investigate the potency of two amphetamines, dextroamphetamine (d-AMPH) and methamphetamine (m-AMPH), on the behavior and energetic dysfunction in the brain of rats. Dextroamphetamine 87-104 amphiphysin Rattus norvegicus 108-112 22957537-2 2012 Given the paucity of information regarding the impact of cholesterol on substrate efflux by the DAT, this study explores its influence on the kinetics of DAT-mediated DA efflux induced by dextroamphetamine, as measured by rotating disk electrode voltammetry (RDEV). Dextroamphetamine 188-205 solute carrier family 6 member 3 Homo sapiens 96-99 22957537-2 2012 Given the paucity of information regarding the impact of cholesterol on substrate efflux by the DAT, this study explores its influence on the kinetics of DAT-mediated DA efflux induced by dextroamphetamine, as measured by rotating disk electrode voltammetry (RDEV). Dextroamphetamine 188-205 solute carrier family 6 member 3 Homo sapiens 154-157 23076832-8 2012 Western blot showed that d-AMPH significantly increased GSK-3 and PKC levels, and decreased pGSK-3, PKA, NGF, BDNF and CREB levels in the structures analyzed. Dextroamphetamine 25-31 protein kinase C, gamma Rattus norvegicus 66-69 23076832-8 2012 Western blot showed that d-AMPH significantly increased GSK-3 and PKC levels, and decreased pGSK-3, PKA, NGF, BDNF and CREB levels in the structures analyzed. Dextroamphetamine 25-31 nerve growth factor Rattus norvegicus 105-108 23076832-8 2012 Western blot showed that d-AMPH significantly increased GSK-3 and PKC levels, and decreased pGSK-3, PKA, NGF, BDNF and CREB levels in the structures analyzed. Dextroamphetamine 25-31 brain-derived neurotrophic factor Rattus norvegicus 110-114 23076832-8 2012 Western blot showed that d-AMPH significantly increased GSK-3 and PKC levels, and decreased pGSK-3, PKA, NGF, BDNF and CREB levels in the structures analyzed. Dextroamphetamine 25-31 cAMP responsive element binding protein 1 Rattus norvegicus 119-123 23076832-10 2012 The present study demonstrated that the PKC inhibitor modulates the alterations in the behavior, neurotrophic and apoptosis pathway induced by d-AMPH, reinforcing the need for more studies of PKC as a possible target for treatment of bipolar disorder. Dextroamphetamine 143-149 protein kinase C, gamma Rattus norvegicus 40-43 23076832-10 2012 The present study demonstrated that the PKC inhibitor modulates the alterations in the behavior, neurotrophic and apoptosis pathway induced by d-AMPH, reinforcing the need for more studies of PKC as a possible target for treatment of bipolar disorder. Dextroamphetamine 143-149 protein kinase C, gamma Rattus norvegicus 192-195 22897629-14 2012 Furthermore, ALA reversed AMPH-induced decreases in BDNF and GSH in the HC. Dextroamphetamine 26-30 brain derived neurotrophic factor Mus musculus 52-56 22796652-6 2012 Our results found that d-amphetamine caused a marked increase in glia fibrillary acidic protein (GFAP), an astroglia marker, expression that implicated astrogliosis in both hippocampus and prefrontal cortex. Dextroamphetamine 23-36 glial fibrillary acidic protein Rattus norvegicus 65-95 22796652-6 2012 Our results found that d-amphetamine caused a marked increase in glia fibrillary acidic protein (GFAP), an astroglia marker, expression that implicated astrogliosis in both hippocampus and prefrontal cortex. Dextroamphetamine 23-36 glial fibrillary acidic protein Rattus norvegicus 97-101 22271821-10 2012 In the DAT efflux experiments, D-amphetamine (full substrate) promoted a fast efflux (K1 = 0.24 min(-1)) and a slow efflux (K2 = 0.008 min(-1)). Dextroamphetamine 31-44 solute carrier family 6 member 3 Rattus norvegicus 7-10 22537773-5 2012 We have previously found that the forced expression of Bcl-X(L) in these cells enhances DAn generation and improves, short-term, d-amphetamine-induced rotation after transplantation in the 6-OH-DA rat model of PD 2-month post-grafting. Dextroamphetamine 129-142 Bcl2-like 1 Rattus norvegicus 55-63 22079347-6 2012 In locomotor activity studies, both single and repeated exposure to d-amphetamine or methamphetamine generated significantly higher levels of total distance traveled in TAAR1 KO mice compared to WT mice. Dextroamphetamine 68-81 trace amine-associated receptor 1 Mus musculus 169-174 22079585-7 2012 Both apomorphine and d-amphetamine increased total distance activity in Adh1-/- mice at both age intervals and in Adh1/4-/- mice at 7 months of age compared to WT mice. Dextroamphetamine 21-34 alcohol dehydrogenase 1 (class I) Mus musculus 72-76 22079585-7 2012 Both apomorphine and d-amphetamine increased total distance activity in Adh1-/- mice at both age intervals and in Adh1/4-/- mice at 7 months of age compared to WT mice. Dextroamphetamine 21-34 alcohol dehydrogenase 1 (class I) Mus musculus 114-120 21316929-1 2011 We have previously shown that both the psychostimulant d-amphetamine and the antipsychotics haloperidol and risperidone affect extracellular concentrations and tissue content of neurotensin (NT) in distinct brain regions. Dextroamphetamine 55-68 neurotensin Rattus norvegicus 178-189 22952603-0 2012 Genome-wide association study of d-amphetamine response in healthy volunteers identifies putative associations, including cadherin 13 (CDH13). Dextroamphetamine 33-46 cadherin 13 Homo sapiens 122-133 22952603-0 2012 Genome-wide association study of d-amphetamine response in healthy volunteers identifies putative associations, including cadherin 13 (CDH13). Dextroamphetamine 33-46 cadherin 13 Homo sapiens 135-140 21307816-8 2011 However, there was a statistically significant left-right asymmetry after intake of the combination promethazine + d-amphetamine for the parameters p13 and latency difference. Dextroamphetamine 115-128 H3 histone pseudogene 6 Homo sapiens 148-151 21354322-3 2011 The enzyme is competitively inhibited by hMAO A selective reversible inhibitors with the exception of d-amphetamine where uncompetitive inhibition is exhibited. Dextroamphetamine 102-115 monoamine oxidase A Homo sapiens 41-47 20813138-4 2011 SD and WKY rats treated with d-amphetamine displayed better STM and LTM, compared to SD-vehicle, WKY-vehicle or SHR-d-amphetamine groups. Dextroamphetamine 29-42 sulfotransferase family 1A member 1 Rattus norvegicus 60-63 21408181-7 2011 In NK1R-/- mice, perseveration in the LITI was increased by injection-stress but reduced by d-amphetamine. Dextroamphetamine 92-105 tachykinin receptor 1 Mus musculus 3-7 21408181-11 2011 Because d-amphetamine reduced perseveration in NK1R-/- mice, this action does not require functional NK1R. Dextroamphetamine 8-21 tachykinin receptor 1 Mus musculus 47-51 21029375-4 2011 Associations between levels of self-reported Euphoria, Energy and Stimulation [Addiction Research Center Inventory 49-item questionnaire (ARCI-49)] after d-amphetamine ingestion and polymorphisms in OPRM1 were investigated. Dextroamphetamine 154-167 opioid receptor mu 1 Homo sapiens 199-204 20580908-3 2010 Recently, it is demonstrated that mecamylamine, a nAChR antagonist blocks cocaine-, d-amphetamine-, ephedrine-, nicotine-, and methylphenidate-induced psychomotor sensitization. Dextroamphetamine 84-97 cholinergic receptor, nicotinic, alpha polypeptide 7 Mus musculus 50-55 21113004-8 2010 d-Amphetamine and dizocilpine disrupted PPI in WT and NTS2(-/-) mice but not in NTS1(-/-) mice. Dextroamphetamine 0-13 nuclear encoded tRNA selenocysteine 2 (anticodon TCA) Mus musculus 54-58 21113004-12 2010 Our results indicate that NTS1 seems essential for d-amphetamine and dizocilpine disruption of PPI. Dextroamphetamine 51-64 neurotensin Mus musculus 26-30 20890227-2 2010 This study aimed to further assess the role of beta2 and coexpressed nAChR subunits in the brain (alpha4, alpha6 and alpha7) to control monoamine-mediated locomotor response, that is, response to novelty, saline, nicotine with tranylcypromine pretreatment, cocaine, d-amphetamine and morphine treatments. Dextroamphetamine 266-279 hemoglobin, beta adult minor chain Mus musculus 47-52 20890227-2 2010 This study aimed to further assess the role of beta2 and coexpressed nAChR subunits in the brain (alpha4, alpha6 and alpha7) to control monoamine-mediated locomotor response, that is, response to novelty, saline, nicotine with tranylcypromine pretreatment, cocaine, d-amphetamine and morphine treatments. Dextroamphetamine 266-279 cholinergic receptor, nicotinic, alpha polypeptide 7 Mus musculus 69-74 20890227-2 2010 This study aimed to further assess the role of beta2 and coexpressed nAChR subunits in the brain (alpha4, alpha6 and alpha7) to control monoamine-mediated locomotor response, that is, response to novelty, saline, nicotine with tranylcypromine pretreatment, cocaine, d-amphetamine and morphine treatments. Dextroamphetamine 266-279 immunoglobulin (CD79A) binding protein 1 Mus musculus 98-123 20626559-1 2010 D-amphetamine (AMPH) down-regulates the norepinephrine transporter (NET), although the exact trafficking pathways altered and motifs involved are not known. Dextroamphetamine 0-13 solute carrier family 6 member 2 Homo sapiens 40-66 20626559-1 2010 D-amphetamine (AMPH) down-regulates the norepinephrine transporter (NET), although the exact trafficking pathways altered and motifs involved are not known. Dextroamphetamine 15-19 solute carrier family 6 member 2 Homo sapiens 40-66 20626559-11 2010 The results demonstrate that reduced plasma membrane insertion and enhanced endocytosis account for AMPH-mediated NET down-regulation, and provide the first evidence that T258/S259 motif is involved only in AMPH-induced NET endocytosis that is desipramine-sensitive, but PKC and CaMKII independent. Dextroamphetamine 207-211 calcium/calmodulin dependent protein kinase II gamma Homo sapiens 279-285 20091113-0 2010 Polymorphisms in dopamine transporter (SLC6A3) are associated with stimulant effects of D-amphetamine: an exploratory pharmacogenetic study using healthy volunteers. Dextroamphetamine 88-101 solute carrier family 6 member 3 Homo sapiens 17-37 20492355-1 2010 Our previous work suggested a role for oligomerization in regulating dopamine transporter (DAT) internalization, with d-amphetamine dissociating DAT oligomers and monomers being endocytosed. Dextroamphetamine 118-131 solute carrier family 6 member 3 Homo sapiens 145-148 20492355-3 2010 Upon pairing wild-type (WT) DAT with W84L mutant, effects of d-amphetamine on oligomerization (decrease) but not surface DAT are observed. Dextroamphetamine 61-74 solute carrier family 6 member 3 Homo sapiens 28-31 20402963-7 2010 Inhibition studies demonstrated that d-AMPH exerts relatively weak inhibitory effects on the OCT3-mediated uptake of DA, NE, 5-HT, and the model OCT3 substrate 4-(4-(dimethylamino)styryl)-N-methylpyridinium iodide. Dextroamphetamine 37-43 solute carrier family 22 (organic cation transporter), member 3 Mus musculus 93-97 20402963-7 2010 Inhibition studies demonstrated that d-AMPH exerts relatively weak inhibitory effects on the OCT3-mediated uptake of DA, NE, 5-HT, and the model OCT3 substrate 4-(4-(dimethylamino)styryl)-N-methylpyridinium iodide. Dextroamphetamine 37-43 solute carrier family 22 (organic cation transporter), member 3 Mus musculus 145-149 20402963-9 2010 Furthermore, the disposition of d-AMPH in various tissues including the brain, liver, heart, kidney, muscle, intestine, spleen, testis, uterus, and plasma were determined in both male and female Oct3 KO and WT mice. Dextroamphetamine 32-38 solute carrier family 22 (organic cation transporter), member 3 Mus musculus 195-199 20193696-5 2010 Systemic administration of d-amphetamine (4 mg/kg, ip) increased glutamate release by 500% in WT mice, as compared to 300% in NTS2(-/-) mice, and 250% in NTS1(-/-) mice. Dextroamphetamine 27-40 nuclear encoded tRNA selenocysteine 2 (anticodon TCA) Mus musculus 126-130 20193696-5 2010 Systemic administration of d-amphetamine (4 mg/kg, ip) increased glutamate release by 500% in WT mice, as compared to 300% in NTS2(-/-) mice, and 250% in NTS1(-/-) mice. Dextroamphetamine 27-40 neurotensin Mus musculus 154-158 20193696-6 2010 Additionally, d-amphetamine injection caused a 4-fold increase in GABA release in both WT and NTS2(-/-) mice, but only a 2-fold increase in NTS1(-/-) mice. Dextroamphetamine 14-27 nuclear encoded tRNA selenocysteine 2 (anticodon TCA) Mus musculus 94-98 20193696-7 2010 Therefore, NTS1 and NTS2 modulate basal release of D-serine and glutamate, and also d-amphetamine-induced GABA and glutamate release in striatum. Dextroamphetamine 84-97 neurotensin Mus musculus 11-15 20193696-7 2010 Therefore, NTS1 and NTS2 modulate basal release of D-serine and glutamate, and also d-amphetamine-induced GABA and glutamate release in striatum. Dextroamphetamine 84-97 nuclear encoded tRNA selenocysteine 2 (anticodon TCA) Mus musculus 20-24 20211191-8 2010 NTS1(-/-) mice showed higher locomotor activity and exaggerated dopamine release in response to d-amphetamine. Dextroamphetamine 96-109 neurotensin Mus musculus 0-4 20091113-0 2010 Polymorphisms in dopamine transporter (SLC6A3) are associated with stimulant effects of D-amphetamine: an exploratory pharmacogenetic study using healthy volunteers. Dextroamphetamine 88-101 solute carrier family 6 member 3 Homo sapiens 39-45 20131069-0 2010 Effects of a gastrin-releasing peptide receptor antagonist on D-amphetamine-induced oxidative stress in the rat brain. Dextroamphetamine 62-75 gastrin releasing peptide receptor Rattus norvegicus 13-47 19958389-7 2010 Upon the clearly symptomatic stage, 5-month-old Mecp2-308 mice were also associated with reduced spontaneous home-cage motor activity, motor coordination impairments (rotarod and dowel tests), and a more marked profile of D-amphetamine (10 mg/kg) released stereotyped behavioral syndrome than wt mice. Dextroamphetamine 222-235 methyl CpG binding protein 2 Mus musculus 48-53 19204064-6 2010 A lack of functional NK1R reduced (>50%) spontaneous dopamine efflux in the prefrontal cortex and abolished the striatal dopamine response to d-amphetamine. Dextroamphetamine 145-158 tachykinin receptor 1 Mus musculus 21-25 19932152-0 2010 A sensitizing D-amphetamine dose regimen induces long-lasting spinophilin and VGLUT1 protein upregulation in the rat diencephalon. Dextroamphetamine 14-27 protein phosphatase 1, regulatory subunit 9B Rattus norvegicus 62-73 19932152-0 2010 A sensitizing D-amphetamine dose regimen induces long-lasting spinophilin and VGLUT1 protein upregulation in the rat diencephalon. Dextroamphetamine 14-27 solute carrier family 17 member 7 Rattus norvegicus 78-84 19932152-7 2010 Strong positive correlations were observed between VGLUT1 and spinophilin expression in PVT, medial habenula, MD, VM and LH of D-AMPH-treated rats. Dextroamphetamine 127-133 solute carrier family 17 member 7 Rattus norvegicus 51-57 19932152-7 2010 Strong positive correlations were observed between VGLUT1 and spinophilin expression in PVT, medial habenula, MD, VM and LH of D-AMPH-treated rats. Dextroamphetamine 127-133 protein phosphatase 1, regulatory subunit 9B Rattus norvegicus 62-73 19748515-4 2009 In the course of this work, we discovered that NK1R-/- mice express locomotor hyperactivity that is prevented by psychostimulants (d-amphetamine or methylphenidate). Dextroamphetamine 131-144 tachykinin receptor 1 Mus musculus 47-51 19968402-0 2009 Evaluation of genetic variability in the dopamine receptor D2 in relation to behavioral inhibition and impulsivity/sensation seeking: an exploratory study with d-amphetamine in healthy participants. Dextroamphetamine 160-173 dopamine receptor D2 Homo sapiens 41-61 19968402-7 2009 We secondarily evaluated the DRD2 SNPs in relation to response to d-amphetamine on stop task performance and mood ratings. Dextroamphetamine 66-79 dopamine receptor D2 Homo sapiens 29-33 19748515-5 2009 Moreover, hyperactivity is induced in wildtypes by treating them with an NK1R antagonist (at doses that have no effect on the behaviour of NK1R-/- mice): this hyperactivity is prevented by d-amphetamine, as in NK1R-/- mice. Dextroamphetamine 189-202 tachykinin receptor 1 Mus musculus 73-77 19615368-7 2009 A protective effect against ipsilateral d-amphetamine-induced turning was seen with 10 microg wtGDNF, 17 microg HB-GAM+10 microg wtGDNF or 10 microg Delta38N-GDNF at 8 weeks post lesion. Dextroamphetamine 40-53 pleiotrophin Rattus norvegicus 112-118 19912621-3 2009 PKCI/HINT1 gene knockout (KO) mice display hyper-locomotion in response to D-amphetamine which is considered a positive symptom of schizophrenia in animal models. Dextroamphetamine 75-88 protein kinase C, iota Mus musculus 0-4 19912621-3 2009 PKCI/HINT1 gene knockout (KO) mice display hyper-locomotion in response to D-amphetamine which is considered a positive symptom of schizophrenia in animal models. Dextroamphetamine 75-88 histidine triad nucleotide binding protein 1 Mus musculus 5-10 19615368-7 2009 A protective effect against ipsilateral d-amphetamine-induced turning was seen with 10 microg wtGDNF, 17 microg HB-GAM+10 microg wtGDNF or 10 microg Delta38N-GDNF at 8 weeks post lesion. Dextroamphetamine 40-53 glial cell derived neurotrophic factor Rattus norvegicus 96-100 19423342-3 2009 Compound 4a, incorporating d-amphetamine, caused significant inhibition of cholinesterase in vivo at doses that were well tolerated. Dextroamphetamine 27-40 butyrylcholinesterase Rattus norvegicus 75-89 19389387-6 2009 While all groups of rats showed a significant reduction in d-amphetamine-induced rotations at 6 weeks posttransplantation a significantly improved graft function was observed only in the days in vitro (DIV) 4 GDNF pretreated group compared to the control group. Dextroamphetamine 59-72 glial cell derived neurotrophic factor Rattus norvegicus 209-213 19727679-1 2009 BACKGROUND AND RATIONALE: We previously found that the intronic norepinephrine transporter gene (SLC6A2) polymorphism rs36017 modulates feelings of elation after administration of 20 mg D-amphetamine in healthy volunteers. Dextroamphetamine 186-199 solute carrier family 6 member 2 Homo sapiens 97-103 19727679-2 2009 OBJECTIVES: In this study, we further investigated the association between D-amphetamine response and 11 SLC6A2 single-nucleotide polymorphisms (SNPs), including rs36017, in an extended sample of Caucasian young adults. Dextroamphetamine 75-88 solute carrier family 6 member 2 Homo sapiens 105-111 19727679-7 2009 CONCLUSIONS: These results provide further evidence that genetic variants in the SLC6A2 gene are involved in acute response to D-amphetamine, which may influence progression to amphetamine abuse. Dextroamphetamine 127-140 solute carrier family 6 member 2 Homo sapiens 81-87 19553455-5 2009 5-HT(2C)R mutant mice displayed increased activity of substantia nigra pars compacta (SNc) dopaminergic neurons, elevated baseline extracellular dopamine concentrations in the dorsal striatum (DSt), alterations in grooming behavior, and enhanced sensitivity to the stereotypic behavioral effects of d-amphetamine and GBR 12909. Dextroamphetamine 299-312 5-hydroxytryptamine (serotonin) receptor 2C Mus musculus 0-7 19519772-3 2009 The potency of p-tyramine and other non-catechols (d-amphetamine, beta-phenethylamine, MPP(+)) in inhibiting cocaine analog binding to DAT in digitonin-treated cells was markedly weakened to a level similar to that observed in cell-free membranes. Dextroamphetamine 51-64 solute carrier family 6 member 3 Homo sapiens 135-138 19581206-0 2009 D-amphetamine toxicity in freshly isolated rat hepatocytes: a possible role of CYP3A. Dextroamphetamine 0-13 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 79-84 19236563-0 2009 Phosphodiesterase 10A inhibition modulates the sensitivity of the mesolimbic dopaminergic system to D-amphetamine: involvement of the D1-regulated feedback control of midbrain dopamine neurons. Dextroamphetamine 100-113 phosphodiesterase 10A Homo sapiens 0-17 19236563-4 2009 We found that the selective PDE10A inhibitor, MP-10, blocked D-amphetamine-induced hyperactivity as well as D-amphetamine-induced dopamine efflux in the nucleus accumbens in a dose-dependent manner. Dextroamphetamine 61-74 phosphodiesterase 10A Homo sapiens 28-34 19236563-4 2009 We found that the selective PDE10A inhibitor, MP-10, blocked D-amphetamine-induced hyperactivity as well as D-amphetamine-induced dopamine efflux in the nucleus accumbens in a dose-dependent manner. Dextroamphetamine 108-121 phosphodiesterase 10A Homo sapiens 28-34 19236563-8 2009 These data suggest that the D(1)-regulated feedback control of midbrain dopamine neurons is a critical pathway involved in the modulation of the response of mesolimbic dopamine neurons to D-amphetamine by PDE10A inhibition. Dextroamphetamine 188-201 phosphodiesterase 10A Homo sapiens 205-211 19050854-2 2009 Subsequently, we identified a single nucleotide polymorphism in CSNK1E (rs135745) that was associated with increased sensitivity to the subjective effects of d-amphetamine in healthy human subjects. Dextroamphetamine 158-171 casein kinase 1 epsilon Homo sapiens 64-70 19244097-5 2009 In the present study, we report that SoRI-9804 and SoRI-20040, at doses that do not alter release, partially inhibited d-amphetamine-induced DAT-mediated release of [(3)H]1-methyl-4-phenylpyridinium (MPP(+))or[(3)H]dopamine from striatal synaptosomes ("DAT-mediated DA release") in a dose-dependent manner. Dextroamphetamine 119-132 solute carrier family 6 member 3 Homo sapiens 141-144 19244097-10 2009 The two major findings of this study are 1) the identification of both "agonist" (SoRI-9804 and SoRI-20040) and "antagonist" (SoRI-20041) allosteric modulators of D-amphetamine-induced DAT-mediated DA release and 2) [(3)H]DA uptake and d-amphetamine-induced DAT-mediated efflux can be separately modulated. Dextroamphetamine 163-176 solute carrier family 6 member 3 Homo sapiens 185-188 19244097-10 2009 The two major findings of this study are 1) the identification of both "agonist" (SoRI-9804 and SoRI-20040) and "antagonist" (SoRI-20041) allosteric modulators of D-amphetamine-induced DAT-mediated DA release and 2) [(3)H]DA uptake and d-amphetamine-induced DAT-mediated efflux can be separately modulated. Dextroamphetamine 163-176 solute carrier family 6 member 3 Homo sapiens 258-261 18424625-10 2008 on PCP and AMP-evoked behavioral activation were absent in mGlu2 and mGlu2/3 but not in mGlu3 receptor-deficient mice, indicating that the activation of mGlu2 and not mGlu3 receptors is responsible for the antipsychotic-like effects of the mGlu2/3 receptor agonist LY404039. Dextroamphetamine 11-14 glutamate receptor, metabotropic 3 Mus musculus 167-172 19116182-0 2009 CB1 receptor knockout mice are hyporesponsive to the behavior-stimulating actions of d-amphetamine: role of mGlu5 receptors. Dextroamphetamine 85-98 cannabinoid receptor 1 (brain) Mus musculus 0-3 19116182-2 2009 The CB1R antagonist SR141716A dose-dependently decreased d-amphetamine-induced hyperactivity.Also, d-amphetamine-induced hyperlocomotion was reduced in CB1R knockout (KO) mice. Dextroamphetamine 57-70 cannabinoid receptor 1 (brain) Mus musculus 4-8 19116182-2 2009 The CB1R antagonist SR141716A dose-dependently decreased d-amphetamine-induced hyperactivity.Also, d-amphetamine-induced hyperlocomotion was reduced in CB1R knockout (KO) mice. Dextroamphetamine 57-70 cannabinoid receptor 1 (brain) Mus musculus 152-156 19116182-2 2009 The CB1R antagonist SR141716A dose-dependently decreased d-amphetamine-induced hyperactivity.Also, d-amphetamine-induced hyperlocomotion was reduced in CB1R knockout (KO) mice. Dextroamphetamine 99-112 cannabinoid receptor 1 (brain) Mus musculus 4-8 19116182-2 2009 The CB1R antagonist SR141716A dose-dependently decreased d-amphetamine-induced hyperactivity.Also, d-amphetamine-induced hyperlocomotion was reduced in CB1R knockout (KO) mice. Dextroamphetamine 99-112 cannabinoid receptor 1 (brain) Mus musculus 152-156 19116182-3 2009 However, CB1R KO and wild-type mice showed a similar d-amphetamine-induced increase in nucleus accumbens DA release. Dextroamphetamine 53-66 cannabinoid receptor 1 (brain) Mus musculus 9-13 19116182-5 2009 Blockade of metabotropic-glutamate-receptors-5 (mGluR5)with MPEP, but not blockade of N-methyl-D-aspartate-receptors (NMDA) with MK-801,restored to a great extent the blunted d-amphetamine-induced hyperlocomotion seen after CB1R antagonism/invalidation. Dextroamphetamine 175-188 glutamate receptor, ionotropic, kainate 1 Mus musculus 48-54 19116182-6 2009 Thus, hyporesponsiveness to the psychostimulant effects of d-amphetamine as a result of CB1R antagonism/invalidation is not due to an ensuing decrease in d-amphetamine-induced DA release in the nucleus accumbens, but rather due to a hyperglutamatergic state and facilitation of glutamatergic neurotransmission at the mGlu5, but not NMDA, receptors. Dextroamphetamine 59-72 cannabinoid receptor 1 (brain) Mus musculus 88-92 19183252-1 2009 The dopamine transporter (DAT) substrates dopamine, d-amphetamine (AMPH), and methamphetamine are known to rapidly and transiently reduce DAT activity and/or surface expression in dorsal striatum and heterologous expression systems. Dextroamphetamine 52-65 solute carrier family 6 member 3 Rattus norvegicus 4-24 19183252-1 2009 The dopamine transporter (DAT) substrates dopamine, d-amphetamine (AMPH), and methamphetamine are known to rapidly and transiently reduce DAT activity and/or surface expression in dorsal striatum and heterologous expression systems. Dextroamphetamine 52-65 solute carrier family 6 member 3 Rattus norvegicus 26-29 19183252-1 2009 The dopamine transporter (DAT) substrates dopamine, d-amphetamine (AMPH), and methamphetamine are known to rapidly and transiently reduce DAT activity and/or surface expression in dorsal striatum and heterologous expression systems. Dextroamphetamine 52-65 solute carrier family 6 member 3 Rattus norvegicus 138-141 19183252-1 2009 The dopamine transporter (DAT) substrates dopamine, d-amphetamine (AMPH), and methamphetamine are known to rapidly and transiently reduce DAT activity and/or surface expression in dorsal striatum and heterologous expression systems. Dextroamphetamine 67-71 solute carrier family 6 member 3 Rattus norvegicus 4-24 19183252-1 2009 The dopamine transporter (DAT) substrates dopamine, d-amphetamine (AMPH), and methamphetamine are known to rapidly and transiently reduce DAT activity and/or surface expression in dorsal striatum and heterologous expression systems. Dextroamphetamine 67-71 solute carrier family 6 member 3 Rattus norvegicus 26-29 19183252-1 2009 The dopamine transporter (DAT) substrates dopamine, d-amphetamine (AMPH), and methamphetamine are known to rapidly and transiently reduce DAT activity and/or surface expression in dorsal striatum and heterologous expression systems. Dextroamphetamine 67-71 solute carrier family 6 member 3 Rattus norvegicus 138-141 19183252-8 2009 Moreover, exposure to AMPH appears to regulate striatal DATs in a biphasic manner, with an initial protein kinase C-dependent decrease in DAT-mediated uptake velocity and then, with longer exposure, a reduction in DAT surface expression. Dextroamphetamine 22-26 solute carrier family 6 member 3 Rattus norvegicus 56-59 19183252-8 2009 Moreover, exposure to AMPH appears to regulate striatal DATs in a biphasic manner, with an initial protein kinase C-dependent decrease in DAT-mediated uptake velocity and then, with longer exposure, a reduction in DAT surface expression. Dextroamphetamine 22-26 solute carrier family 6 member 3 Rattus norvegicus 138-141 19222557-3 2009 Here, for the first time, the effect of methylphenidate (MPH) and D-amphetamine (AMPH) on the expression of two key genes for neuronal development and plasticity, brain-derived neurotrophic factor (bdnf) and the effector immediate early gene activity-regulated, cytoskeletal-associated protein (Arc), was examined in both juvenile and adult rats. Dextroamphetamine 66-79 brain-derived neurotrophic factor Rattus norvegicus 163-196 19222557-3 2009 Here, for the first time, the effect of methylphenidate (MPH) and D-amphetamine (AMPH) on the expression of two key genes for neuronal development and plasticity, brain-derived neurotrophic factor (bdnf) and the effector immediate early gene activity-regulated, cytoskeletal-associated protein (Arc), was examined in both juvenile and adult rats. Dextroamphetamine 66-79 brain-derived neurotrophic factor Rattus norvegicus 198-202 19222557-3 2009 Here, for the first time, the effect of methylphenidate (MPH) and D-amphetamine (AMPH) on the expression of two key genes for neuronal development and plasticity, brain-derived neurotrophic factor (bdnf) and the effector immediate early gene activity-regulated, cytoskeletal-associated protein (Arc), was examined in both juvenile and adult rats. Dextroamphetamine 66-79 activity-regulated cytoskeleton-associated protein Rattus norvegicus 242-293 19222557-3 2009 Here, for the first time, the effect of methylphenidate (MPH) and D-amphetamine (AMPH) on the expression of two key genes for neuronal development and plasticity, brain-derived neurotrophic factor (bdnf) and the effector immediate early gene activity-regulated, cytoskeletal-associated protein (Arc), was examined in both juvenile and adult rats. Dextroamphetamine 66-79 activity-regulated cytoskeleton-associated protein Rattus norvegicus 295-298 19222557-3 2009 Here, for the first time, the effect of methylphenidate (MPH) and D-amphetamine (AMPH) on the expression of two key genes for neuronal development and plasticity, brain-derived neurotrophic factor (bdnf) and the effector immediate early gene activity-regulated, cytoskeletal-associated protein (Arc), was examined in both juvenile and adult rats. Dextroamphetamine 81-85 brain-derived neurotrophic factor Rattus norvegicus 163-196 19222557-3 2009 Here, for the first time, the effect of methylphenidate (MPH) and D-amphetamine (AMPH) on the expression of two key genes for neuronal development and plasticity, brain-derived neurotrophic factor (bdnf) and the effector immediate early gene activity-regulated, cytoskeletal-associated protein (Arc), was examined in both juvenile and adult rats. Dextroamphetamine 81-85 brain-derived neurotrophic factor Rattus norvegicus 198-202 19222557-3 2009 Here, for the first time, the effect of methylphenidate (MPH) and D-amphetamine (AMPH) on the expression of two key genes for neuronal development and plasticity, brain-derived neurotrophic factor (bdnf) and the effector immediate early gene activity-regulated, cytoskeletal-associated protein (Arc), was examined in both juvenile and adult rats. Dextroamphetamine 81-85 activity-regulated cytoskeleton-associated protein Rattus norvegicus 242-293 19222557-3 2009 Here, for the first time, the effect of methylphenidate (MPH) and D-amphetamine (AMPH) on the expression of two key genes for neuronal development and plasticity, brain-derived neurotrophic factor (bdnf) and the effector immediate early gene activity-regulated, cytoskeletal-associated protein (Arc), was examined in both juvenile and adult rats. Dextroamphetamine 81-85 activity-regulated cytoskeleton-associated protein Rattus norvegicus 295-298 19047053-4 2009 In the in vitro expression system, DAT-mediated whole-cell currents were greater for METH stimulation than for AMPH. Dextroamphetamine 111-115 solute carrier family 6 member 3 Rattus norvegicus 35-38 19047053-8 2009 Intact phosphorylation sites in the N-terminal domain of DAT were required for the AMPH- and METH-induced increase in [Ca(2+)](i) and for the enhanced effects of METH on [Ca(2+)](i) elevation. Dextroamphetamine 83-87 solute carrier family 6 member 3 Rattus norvegicus 57-60 19047053-11 2009 Together these data demonstrate that METH has a stronger effect on DAT-mediated cell physiology than AMPH, which may contribute to the euphoric and addictive properties of METH compared with AMPH. Dextroamphetamine 191-195 solute carrier family 6 member 3 Rattus norvegicus 67-70 19046390-0 2008 A sensitizing d-amphetamine regimen induces long-lasting spinophilin protein upregulation in the rat striatum and limbic forebrain. Dextroamphetamine 14-27 protein phosphatase 1, regulatory subunit 9B Rattus norvegicus 57-68 18991853-0 2008 Transient enhanced expression of Cdk5 activator p25 after acute and chronic d-amphetamine administration. Dextroamphetamine 76-89 cyclin-dependent kinase 5 Rattus norvegicus 33-37 18991853-0 2008 Transient enhanced expression of Cdk5 activator p25 after acute and chronic d-amphetamine administration. Dextroamphetamine 76-89 lipocalin 2 Rattus norvegicus 48-51 18991853-7 2008 In this study we report that the cdk5/p35 complex participates in acute and chronic d-amphetamine (AMPH)-evoked behavioral events, and we show a surprisingly transient enhanced expression of p25 and a lasting increased expression of p35 in dorsal striatal synaptosomes after acute and chronic AMPH administration. Dextroamphetamine 84-97 cyclin-dependent kinase 5 Rattus norvegicus 33-37 18991853-7 2008 In this study we report that the cdk5/p35 complex participates in acute and chronic d-amphetamine (AMPH)-evoked behavioral events, and we show a surprisingly transient enhanced expression of p25 and a lasting increased expression of p35 in dorsal striatal synaptosomes after acute and chronic AMPH administration. Dextroamphetamine 84-97 cyclin-dependent kinase 5 regulatory subunit 1 Rattus norvegicus 38-41 18991853-7 2008 In this study we report that the cdk5/p35 complex participates in acute and chronic d-amphetamine (AMPH)-evoked behavioral events, and we show a surprisingly transient enhanced expression of p25 and a lasting increased expression of p35 in dorsal striatal synaptosomes after acute and chronic AMPH administration. Dextroamphetamine 84-97 lipocalin 2 Rattus norvegicus 191-194 18991853-7 2008 In this study we report that the cdk5/p35 complex participates in acute and chronic d-amphetamine (AMPH)-evoked behavioral events, and we show a surprisingly transient enhanced expression of p25 and a lasting increased expression of p35 in dorsal striatal synaptosomes after acute and chronic AMPH administration. Dextroamphetamine 84-97 cyclin-dependent kinase 5 regulatory subunit 1 Rattus norvegicus 233-236 18991853-7 2008 In this study we report that the cdk5/p35 complex participates in acute and chronic d-amphetamine (AMPH)-evoked behavioral events, and we show a surprisingly transient enhanced expression of p25 and a lasting increased expression of p35 in dorsal striatal synaptosomes after acute and chronic AMPH administration. Dextroamphetamine 99-103 cyclin-dependent kinase 5 Rattus norvegicus 33-37 18991853-7 2008 In this study we report that the cdk5/p35 complex participates in acute and chronic d-amphetamine (AMPH)-evoked behavioral events, and we show a surprisingly transient enhanced expression of p25 and a lasting increased expression of p35 in dorsal striatal synaptosomes after acute and chronic AMPH administration. Dextroamphetamine 99-103 cyclin-dependent kinase 5 regulatory subunit 1 Rattus norvegicus 38-41 18991853-7 2008 In this study we report that the cdk5/p35 complex participates in acute and chronic d-amphetamine (AMPH)-evoked behavioral events, and we show a surprisingly transient enhanced expression of p25 and a lasting increased expression of p35 in dorsal striatal synaptosomes after acute and chronic AMPH administration. Dextroamphetamine 293-297 cyclin-dependent kinase 5 Rattus norvegicus 33-37 18991853-7 2008 In this study we report that the cdk5/p35 complex participates in acute and chronic d-amphetamine (AMPH)-evoked behavioral events, and we show a surprisingly transient enhanced expression of p25 and a lasting increased expression of p35 in dorsal striatal synaptosomes after acute and chronic AMPH administration. Dextroamphetamine 293-297 cyclin-dependent kinase 5 regulatory subunit 1 Rattus norvegicus 38-41 18991853-8 2008 Pak1, a substrate for cdk5, is also enriched in the synaptosomal fraction of acute AMPH-treated rats. Dextroamphetamine 83-87 p21 (RAC1) activated kinase 1 Rattus norvegicus 0-4 18991853-8 2008 Pak1, a substrate for cdk5, is also enriched in the synaptosomal fraction of acute AMPH-treated rats. Dextroamphetamine 83-87 cyclin-dependent kinase 5 Rattus norvegicus 22-26 18811678-9 2009 In addition, the NE transporter (NET) inhibitor atomoxetine attenuates some of d-amphetamine"s subjective and physiological effects in humans. Dextroamphetamine 79-92 solute carrier family 6 member 2 Homo sapiens 17-31 18811678-9 2009 In addition, the NE transporter (NET) inhibitor atomoxetine attenuates some of d-amphetamine"s subjective and physiological effects in humans. Dextroamphetamine 79-92 solute carrier family 6 member 2 Homo sapiens 33-36 19116947-0 2009 Differential regulation of prodynophin, c-fos, and serotonin transporter mRNA following withdrawal from a chronic, escalating dose regimen of D-amphetamine. Dextroamphetamine 142-155 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 40-45 19116947-0 2009 Differential regulation of prodynophin, c-fos, and serotonin transporter mRNA following withdrawal from a chronic, escalating dose regimen of D-amphetamine. Dextroamphetamine 142-155 solute carrier family 6 member 4 Rattus norvegicus 51-72 19116947-4 2009 This study examined the effects of chronic, escalating doses of D-AMPH followed by 24 h of withdrawal on the expression of prodynorphin (PD) and c-fos mRNA in limbic regions of the brain, caudate putamen (CPu), and brainstem and SERT mRNA expression in the dorsal raphe nucleus (DRN). Dextroamphetamine 64-70 prodynorphin Rattus norvegicus 123-135 19116947-4 2009 This study examined the effects of chronic, escalating doses of D-AMPH followed by 24 h of withdrawal on the expression of prodynorphin (PD) and c-fos mRNA in limbic regions of the brain, caudate putamen (CPu), and brainstem and SERT mRNA expression in the dorsal raphe nucleus (DRN). Dextroamphetamine 64-70 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-150 19047053-3 2009 Here we compare structurally similar AMPH and METH on DAT function in a heterologous expression system and in an animal model. Dextroamphetamine 37-41 solute carrier family 6 member 3 Rattus norvegicus 54-57 18351285-0 2008 Immunohistochemical increase in cyclooxygenase-2 without apoptosis in different brain areas of subchronic nicotine- and D-amphetamine-treated rats. Dextroamphetamine 120-133 prostaglandin-endoperoxide synthase 2 Rattus norvegicus 32-48 18571730-1 2008 Effects of d-amphetamine on the renal outer medullary potassium (ROMK1) channels were tested in the Xenopus oocytes expression system. Dextroamphetamine 11-24 potassium inwardly rectifying channel subfamily J member 1 S homeolog Xenopus laevis 65-70 17996928-0 2008 Deletion of Melanin-Concentrating Hormone Receptor-1 gene accentuates D-amphetamine-induced psychomotor activation but neither the subsequent development of sensitization nor the expression of conditioned activity in mice. Dextroamphetamine 70-83 melanin-concentrating hormone receptor 1 Mus musculus 12-52 18479829-8 2008 Exercise or AMPH alone significantly increased BDNF protein in sham and CCI rats, but this effect was lost with the combined treatment. Dextroamphetamine 12-16 brain-derived neurotrophic factor Rattus norvegicus 47-51 18479829-9 2008 In sham-injured rats synapsin I increased significantly after AMPH or exercise, but did not increase after combined treatment. Dextroamphetamine 62-66 synapsin I Rattus norvegicus 21-31 17805311-7 2008 Moreover, we show that the effects of SCH23390, a D1 receptor antagonist known to inhibit development of d-amphetamine behavioral sensitization, are due to its 5-HT2C receptor agonist property. Dextroamphetamine 105-118 5-hydroxytryptamine receptor 2C Homo sapiens 160-166 17805311-8 2008 SCH23390 blocks amphetamine-induced release of norepinephrine and RS102221, a 5-HT2C antagonist, can reverse this inhibition as well as inhibition of noradrenergic sensitization and development of behavioral sensitization induced by repeated d-amphetamine. Dextroamphetamine 242-255 5-hydroxytryptamine receptor 2C Homo sapiens 78-84 18571730-4 2008 d-Amphetamine inhibited the activity of ROMK1 channels in a manner that was concentration-dependent but voltage-independent. Dextroamphetamine 0-13 potassium inwardly rectifying channel subfamily J member 1 S homeolog Xenopus laevis 40-45 18571730-8 2008 These findings suggest that d-amphetamine inhibits ROMK1 channels independently of the pH i. Dextroamphetamine 28-41 potassium inwardly rectifying channel subfamily J member 1 S homeolog Xenopus laevis 51-56 18571730-9 2008 The effects of d-amphetamine on ROMK1 channels may be due to a conformational change induced by PKA-mediated phosphorylation, but not to charge-charge interactions. Dextroamphetamine 15-28 potassium inwardly rectifying channel subfamily J member 1 S homeolog Xenopus laevis 32-37 17716785-8 2008 The pro-oxidative effect induced by AMPH and MDMA showed a strong dependence on calcium (extracellular and from internal stores) and also was inhibited by nicotinic receptor (nAChR) antagonists dihydro-beta-erythroidine, methyllycaconitine (MLA) and alpha-bungarotoxin. Dextroamphetamine 36-40 cholinergic receptor, nicotinic, alpha polypeptide 7 Mus musculus 175-180 18000809-6 2008 d-Amphetamine (3 mg/kg) increased c-fos expression within the frontal cortex in MCH(1) receptor KO mice, but not WT mice. Dextroamphetamine 0-13 FBJ osteosarcoma oncogene Mus musculus 34-39 17996928-1 2008 The present study aimed to test the hypothesis that mice lacking the MCHR1 receptor (Melanin-Concentrating Hormone Receptor-1) present an elevated vulnerability towards the neurobehavioural effects of D-amphetamine, presumably due to previously established up-regulations of dopamine D1 receptors in these mice. Dextroamphetamine 201-214 melanin-concentrating hormone receptor 1 Mus musculus 69-74 17996928-1 2008 The present study aimed to test the hypothesis that mice lacking the MCHR1 receptor (Melanin-Concentrating Hormone Receptor-1) present an elevated vulnerability towards the neurobehavioural effects of D-amphetamine, presumably due to previously established up-regulations of dopamine D1 receptors in these mice. Dextroamphetamine 201-214 melanin-concentrating hormone receptor 1 Mus musculus 85-125 17299510-7 2007 Further, we show that although acute administration of lithium and overexpression of the beta-catenin transgene inhibits d-amphetamine-induced hyperlocomotion, neither lithium nor the beta-catenin transgene prevents d-amphetamine-induced sensitization, as measured by locomotor activity. Dextroamphetamine 121-134 catenin (cadherin associated protein), beta 1 Mus musculus 89-101 18250960-10 2008 Further investigation considering sub-sample analysis regarding response to D-amphetamine could enlight the role of SNAP25 in ADHD. Dextroamphetamine 76-89 synaptosome associated protein 25 Homo sapiens 116-122 17681816-2 2007 HYPOTHESIS: Arachidonic acid (AA) signaling via post-synaptic D2 receptors coupled to cytosolic phospholipase A2 (cPLA2) will be reduced in terminal areas ipsilateral to a chronic unilateral substantia nigra lesion in rats given D-amphetamine, which reverses the direction of the DAT, but will be increased in rats given quinpirole, a D2-receptor agonist. Dextroamphetamine 229-242 phospholipase A2 group IVA Rattus norvegicus 86-112 17681816-2 2007 HYPOTHESIS: Arachidonic acid (AA) signaling via post-synaptic D2 receptors coupled to cytosolic phospholipase A2 (cPLA2) will be reduced in terminal areas ipsilateral to a chronic unilateral substantia nigra lesion in rats given D-amphetamine, which reverses the direction of the DAT, but will be increased in rats given quinpirole, a D2-receptor agonist. Dextroamphetamine 229-242 phospholipase A2 group IVA Rattus norvegicus 114-119 17299510-8 2007 Both lithium-treated and beta-catenin mice had an elevated response to d-amphetamine following multiple administrations of the stimulant, though the difference in absolute locomotion was maintained throughout the sensitization time-course. Dextroamphetamine 71-84 catenin (cadherin associated protein), beta 1 Mus musculus 25-37 17614837-5 2007 The present study was conducted to investigate the protective effects of melatonin on d-amphetamine (AMPH)-induced neurotoxicity in cultured human dopaminergic neuroblastoma SK-N-SH cells. Dextroamphetamine 86-99 hedgehog acyltransferase Homo sapiens 174-178 17510759-8 2007 One milligrams per kilogram of prazosin completely blocks d-amphetamine-induced locomotor response in 5-HT2A-R KO naive animals but 3 mg/kg is necessary in sensitized 5-HT2A-R KO mice. Dextroamphetamine 58-71 5-hydroxytryptamine (serotonin) receptor 2A Mus musculus 102-108 17510759-9 2007 CONCLUSIONS: Because naive 5-HT2A-R KO mice exhibit an increased cortical noradrenergic response to d-amphetamine, our data suggest that repeated d-amphetamine modifies noradrenergic transmission in 5-HT2A-R KO mice. Dextroamphetamine 100-113 5-hydroxytryptamine (serotonin) receptor 2A Mus musculus 27-33 17510759-9 2007 CONCLUSIONS: Because naive 5-HT2A-R KO mice exhibit an increased cortical noradrenergic response to d-amphetamine, our data suggest that repeated d-amphetamine modifies noradrenergic transmission in 5-HT2A-R KO mice. Dextroamphetamine 146-159 5-hydroxytryptamine (serotonin) receptor 2A Mus musculus 27-33 17510759-9 2007 CONCLUSIONS: Because naive 5-HT2A-R KO mice exhibit an increased cortical noradrenergic response to d-amphetamine, our data suggest that repeated d-amphetamine modifies noradrenergic transmission in 5-HT2A-R KO mice. Dextroamphetamine 146-159 5-hydroxytryptamine (serotonin) receptor 2A Mus musculus 199-205 17510759-11 2007 Dramatic decrease in SR46349B efficiency in sensitized WT mice indicates that a disruption of the regulating role of 5-HT2A receptors on noradrenergic transmission occurs during sensitization and thus represents the physiological basis of behavioral sensitization to d-amphetamine. Dextroamphetamine 267-280 5-hydroxytryptamine (serotonin) receptor 2A Mus musculus 117-123 17510759-1 2007 RATIONALE: Although locomotor response to d-amphetamine is considered as mediated by an increased release of dopamine in the ventral striatum, blockade of either alpha1b-adrenergic or 5-HT2A receptors almost completely inhibits d-amphetamine-induced locomotor response in mice. Dextroamphetamine 42-55 5-hydroxytryptamine (serotonin) receptor 2A Mus musculus 184-190 17510759-1 2007 RATIONALE: Although locomotor response to d-amphetamine is considered as mediated by an increased release of dopamine in the ventral striatum, blockade of either alpha1b-adrenergic or 5-HT2A receptors almost completely inhibits d-amphetamine-induced locomotor response in mice. Dextroamphetamine 228-241 5-hydroxytryptamine (serotonin) receptor 2A Mus musculus 184-190 17510759-3 2007 However, we show here that, paradoxically, mice lacking 5-HT2A receptors (5-HT2A-R KO) exhibit a twofold higher locomotor response to d-amphetamine than wild-type (WT) littermates. Dextroamphetamine 134-147 5-hydroxytryptamine (serotonin) receptor 2A Mus musculus 56-62 17510759-3 2007 However, we show here that, paradoxically, mice lacking 5-HT2A receptors (5-HT2A-R KO) exhibit a twofold higher locomotor response to d-amphetamine than wild-type (WT) littermates. Dextroamphetamine 134-147 5-hydroxytryptamine (serotonin) receptor 2A Mus musculus 74-80 17510759-6 2007 RESULTS: Repeating amphetamine injections still increases 5-HT2A-R KO mice locomotor response to d-amphetamine at a level similar to that of sensitized WT mice. Dextroamphetamine 97-110 5-hydroxytryptamine (serotonin) receptor 2A Mus musculus 58-64 17614837-5 2007 The present study was conducted to investigate the protective effects of melatonin on d-amphetamine (AMPH)-induced neurotoxicity in cultured human dopaminergic neuroblastoma SK-N-SH cells. Dextroamphetamine 101-105 hedgehog acyltransferase Homo sapiens 174-178 17614837-6 2007 Our data indicate that AMPH significantly reduces cell viability, induces oxidative stress (enhances ROS production and malondialdehyde levels), up-regulates alpha-synuclein expression and decreases intracellular ATP levels. Dextroamphetamine 23-27 synuclein alpha Homo sapiens 158-173 17614837-7 2007 However, pretreatment of SK-N-SH cells with melatonin prevents AMPH-induced loss of cell viability and induction of oxidative stress, while reducing alpha-synuclein expression and increasing ATP production. Dextroamphetamine 63-67 hedgehog acyltransferase Homo sapiens 25-29 16524702-0 2006 Effects of acute and subchronic d-amphetamine on ventral striatal concentrations of neurotensin and neuropeptide Y in rats treated with antipsychotic drugs. Dextroamphetamine 32-45 neurotensin Rattus norvegicus 84-95 17442525-0 2007 Pharmacological modulation of functional connectivity: the correlation structure underlying the phMRI response to d-amphetamine modified by selective dopamine D3 receptor antagonist SB277011A. Dextroamphetamine 114-127 dopamine receptor D3 Rattus norvegicus 150-170 17442525-6 2007 Specifically, we show a modulation of the correlation structure of a probe compound (d-amphetamine) by pretreatment with the selective dopamine D3 receptor antagonist SB277011A in the rat. Dextroamphetamine 85-98 dopamine receptor D3 Rattus norvegicus 135-155 17596442-5 2007 Locomotor responses to cocaine and D-amphetamine were decreased in CB1-R-deficient mice, and sensitization was impaired. Dextroamphetamine 35-48 cannabinoid receptor 1 (brain) Mus musculus 67-70 17239355-0 2007 Norepinephrine transporter gene variation modulates acute response to D-amphetamine. Dextroamphetamine 70-83 solute carrier family 6 member 2 Homo sapiens 0-26 17239355-10 2007 CONCLUSIONS: Polymorphisms in the SLC6A2 gene were associated with mood responses to D-amphetamine. Dextroamphetamine 85-98 solute carrier family 6 member 2 Homo sapiens 34-40 17629354-0 2007 A gastrin-releasing peptide receptor antagonist blocks D-amphetamine-induced hyperlocomotion and increases hippocampal NGF and BDNF levels in rats. Dextroamphetamine 55-68 gastrin releasing peptide receptor Rattus norvegicus 2-36 17374745-0 2007 The acetylcholinesterase inhibitor galantamine inhibits d-amphetamine-induced psychotic-like behavior in Cebus monkeys. Dextroamphetamine 56-69 acetylcholinesterase (Cartwright blood group) Homo sapiens 4-24 17063155-4 2007 Dopamine-stimulated cAMP production in vitro and phosphorylation of AMPA receptor GluR1 subunit in response to D-amph in vivo were decreased in Gnal+/-, but not Drd1a+/- mice. Dextroamphetamine 111-117 glutamate receptor, ionotropic, AMPA1 (alpha 1) Mus musculus 82-87 17063155-4 2007 Dopamine-stimulated cAMP production in vitro and phosphorylation of AMPA receptor GluR1 subunit in response to D-amph in vivo were decreased in Gnal+/-, but not Drd1a+/- mice. Dextroamphetamine 111-117 guanine nucleotide binding protein, alpha stimulating, olfactory type Mus musculus 144-148 17063155-5 2007 Acute locomotor responses to D1 agonist SKF81259, D-amph and cocaine were altered in Gnal+/- mice, and not in Drd1a+/- mice. Dextroamphetamine 50-56 guanine nucleotide binding protein, alpha stimulating, olfactory type Mus musculus 85-89 17063155-7 2007 Gnal+/- mice developed pronounced locomotor sensitization and conditioned locomotor responses after repeated injections of D-amph (2 mg/kg) or cocaine (20 mg/kg). Dextroamphetamine 123-129 guanine nucleotide binding protein, alpha stimulating, olfactory type Mus musculus 0-4 16524702-1 2006 We have reported that acute d-amphetamine increases extracellular concentrations (efflux) of neurotensin-like immunoreactivity (NT-LI) and neuropeptide Y-LI (NPY-LI) in the ventral striatum (VSTR) of freely moving rats, effects that are abolished by chronic administration of haloperidol and risperidone admixed to food pellets. Dextroamphetamine 28-41 neurotensin Rattus norvegicus 93-104 16632196-3 2006 To assess the role of NT in psychostimulant responses, we examined the ability of d-amphetamine (AMP) to induce Fos in wild-type and NT null mutant mice. Dextroamphetamine 97-100 FBJ osteosarcoma oncogene Mus musculus 112-115 16511499-14 2006 These results indicate that D-amphetamine can indirectly activate brain PLA(2) in the unanesthetized rat, and that activation is initiated entirely at D(2)-like receptors. Dextroamphetamine 28-41 phospholipase A2 group IB Rattus norvegicus 72-78 16966188-1 2006 The authors have previously shown an effect of dopamine transporter genotype on acute subjective responses to d-amphetamine, which may affect risk of addiction. Dextroamphetamine 110-123 solute carrier family 6 member 3 Homo sapiens 47-67 16966188-3 2006 The separate and combined analyses of the gene-linked polymorphic region (5-HTTLPR) and the Intron 2 VNTR suggest that these two HTT polymorphisms may contribute to acute subjective responses to d-amphetamine with a small effect. Dextroamphetamine 195-208 huntingtin Homo sapiens 76-79 16632196-3 2006 To assess the role of NT in psychostimulant responses, we examined the ability of d-amphetamine (AMP) to induce Fos in wild-type and NT null mutant mice. Dextroamphetamine 82-95 FBJ osteosarcoma oncogene Mus musculus 112-115 16632196-4 2006 AMP-elicited Fos expression was significantly attenuated in the medial striatum of NT null mutant mice, but was unaffected in other striatal territories. Dextroamphetamine 0-3 FBJ osteosarcoma oncogene Mus musculus 13-16 16632196-4 2006 AMP-elicited Fos expression was significantly attenuated in the medial striatum of NT null mutant mice, but was unaffected in other striatal territories. Dextroamphetamine 0-3 neurotensin Mus musculus 83-85 16632196-7 2006 These data suggest that NT is required for the full activation by AMP of medial striatal neurons. Dextroamphetamine 66-69 neurotensin Mus musculus 24-26 16648258-8 2006 Third, knockout mice for alpha1b-adrenergic (alpha1b-AR KO) or 5-HT(2A) (5-HT(2A)-R KO) receptor, respectively, exhibit a behavioral and biochemical hyperreactivity to the acute injection of p-chloroamphetamine (alpha1b-AR KO; 5-HT levels) and d-amphetamine (5-HT(2A)-R KO; NE levels). Dextroamphetamine 244-257 calcium channel, voltage-dependent, N type, alpha 1B subunit Mus musculus 25-32 16621932-0 2006 The role of the polymorphic efflux transporter P-glycoprotein on the brain accumulation of d-methylphenidate and d-amphetamine. Dextroamphetamine 113-126 phosphoglycolate phosphatase Mus musculus 47-61 16460767-8 2006 In the second experiment, AMPH decreased BDNF levels and Li and VPT increased BDNF levels in rat hippocampus. Dextroamphetamine 26-30 brain-derived neurotrophic factor Rattus norvegicus 41-45 16914949-7 2006 Lithium increased nerve growth factor content in rat hippocampus in both experiments, but this effect was blocked with the co-administration of D-amphetamine. Dextroamphetamine 144-157 nerve growth factor Rattus norvegicus 18-37 16648258-8 2006 Third, knockout mice for alpha1b-adrenergic (alpha1b-AR KO) or 5-HT(2A) (5-HT(2A)-R KO) receptor, respectively, exhibit a behavioral and biochemical hyperreactivity to the acute injection of p-chloroamphetamine (alpha1b-AR KO; 5-HT levels) and d-amphetamine (5-HT(2A)-R KO; NE levels). Dextroamphetamine 244-257 calcium channel, voltage-dependent, N type, alpha 1B subunit Mus musculus 45-52 16648258-8 2006 Third, knockout mice for alpha1b-adrenergic (alpha1b-AR KO) or 5-HT(2A) (5-HT(2A)-R KO) receptor, respectively, exhibit a behavioral and biochemical hyperreactivity to the acute injection of p-chloroamphetamine (alpha1b-AR KO; 5-HT levels) and d-amphetamine (5-HT(2A)-R KO; NE levels). Dextroamphetamine 244-257 calcium channel, voltage-dependent, N type, alpha 1B subunit Mus musculus 45-52 16606363-5 2006 Following D-amphetamine administration, GABAB1-/- mice released less dopamine than wild-type mice, indicative of a reduced cytoplasmic dopamine pool. Dextroamphetamine 10-23 gamma-aminobutyric acid (GABA) B receptor, 1 Mus musculus 40-46 16600190-0 2006 Effects of central leptin infusion on the reward-potentiating effect of D-amphetamine. Dextroamphetamine 72-85 leptin Rattus norvegicus 19-25 16600190-9 2006 leptin infusion in ad libitum fed rats decreased food intake and body weight and enhanced the rewarding effect of D-amphetamine. Dextroamphetamine 114-127 leptin Rattus norvegicus 0-6 16600190-10 2006 Sensitivity to D-amphetamine returned to normal as body weight recovered following cessation of leptin infusion. Dextroamphetamine 15-28 leptin Rattus norvegicus 96-102 16237383-0 2006 Association between the casein kinase 1 epsilon gene region and subjective response to D-amphetamine. Dextroamphetamine 87-100 casein kinase 1, epsilon Mus musculus 24-47 16237383-4 2006 We used a double-blind, crossover design in healthy human volunteers to test association between polymorphisms in the CSNK1E region and subjective response to placebo, 10, or 20 mg of oral D-amphetamine. Dextroamphetamine 189-202 casein kinase 1 epsilon Homo sapiens 118-124 16088329-2 2005 This strain is hemizygous for the SNAP25 gene and displays hyperactivity that responds to dextroamphetamine, but not to methylphenidate. Dextroamphetamine 90-107 synaptosomal-associated protein 25 Mus musculus 34-40 16470869-0 2006 Genetic identification of AChE as a positive modulator of addiction to the psychostimulant D-amphetamine in zebrafish. Dextroamphetamine 91-104 acetylcholinesterase Danio rerio 26-30 16631090-3 2006 We therefore examined if repeated administration of D-AMPH or methamphetamine (METH) may induce loss of binding to striatal DATs in rats by using an experimental biodistribution study design and a SPECT tracer for the DAT ([123I]FP-CIT). Dextroamphetamine 52-58 solute carrier family 6 member 3 Rattus norvegicus 124-127 16122767-1 2005 In heterologous cells expressing the dopamine transporter (DAT), simultaneous elevation of intracellular Na(+) and depolarization of the membrane with gramicidin reduced the potency of various DAT substrates, including dopamine, d-amphetamine, beta-phenethylamine, p-tyramine, and MPP(+), in inhibiting binding of the cocaine analog [(3)H]CFT, with the greatest reduction observed for d-amphetamine. Dextroamphetamine 385-398 solute carrier family 6 member 3 Rattus norvegicus 193-196 15900317-10 2005 By contrast, d-AMPH-treated animals showed marked depletions in striatal DAT and cortical neurodegeneration, but HC and pRh SERT were unaffected. Dextroamphetamine 13-19 solute carrier family 6 member 3 Rattus norvegicus 73-76 16122767-1 2005 In heterologous cells expressing the dopamine transporter (DAT), simultaneous elevation of intracellular Na(+) and depolarization of the membrane with gramicidin reduced the potency of various DAT substrates, including dopamine, d-amphetamine, beta-phenethylamine, p-tyramine, and MPP(+), in inhibiting binding of the cocaine analog [(3)H]CFT, with the greatest reduction observed for d-amphetamine. Dextroamphetamine 229-242 solute carrier family 6 member 3 Rattus norvegicus 37-57 16122767-1 2005 In heterologous cells expressing the dopamine transporter (DAT), simultaneous elevation of intracellular Na(+) and depolarization of the membrane with gramicidin reduced the potency of various DAT substrates, including dopamine, d-amphetamine, beta-phenethylamine, p-tyramine, and MPP(+), in inhibiting binding of the cocaine analog [(3)H]CFT, with the greatest reduction observed for d-amphetamine. Dextroamphetamine 229-242 solute carrier family 6 member 3 Rattus norvegicus 59-62 16122767-1 2005 In heterologous cells expressing the dopamine transporter (DAT), simultaneous elevation of intracellular Na(+) and depolarization of the membrane with gramicidin reduced the potency of various DAT substrates, including dopamine, d-amphetamine, beta-phenethylamine, p-tyramine, and MPP(+), in inhibiting binding of the cocaine analog [(3)H]CFT, with the greatest reduction observed for d-amphetamine. Dextroamphetamine 229-242 solute carrier family 6 member 3 Rattus norvegicus 193-196 16122767-1 2005 In heterologous cells expressing the dopamine transporter (DAT), simultaneous elevation of intracellular Na(+) and depolarization of the membrane with gramicidin reduced the potency of various DAT substrates, including dopamine, d-amphetamine, beta-phenethylamine, p-tyramine, and MPP(+), in inhibiting binding of the cocaine analog [(3)H]CFT, with the greatest reduction observed for d-amphetamine. Dextroamphetamine 385-398 solute carrier family 6 member 3 Rattus norvegicus 37-57 16122767-1 2005 In heterologous cells expressing the dopamine transporter (DAT), simultaneous elevation of intracellular Na(+) and depolarization of the membrane with gramicidin reduced the potency of various DAT substrates, including dopamine, d-amphetamine, beta-phenethylamine, p-tyramine, and MPP(+), in inhibiting binding of the cocaine analog [(3)H]CFT, with the greatest reduction observed for d-amphetamine. Dextroamphetamine 385-398 solute carrier family 6 member 3 Rattus norvegicus 59-62 16122767-2 2005 In rat striatal synaptosomes, gramicidin exerted similar effects; in addition, the potency of d-amphetamine was reduced by the Na(+)-channel activator veratridine. Dextroamphetamine 94-107 sodium voltage-gated channel alpha subunit 8 Rattus norvegicus 127-140