Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Front Cell Neurosci ; 15: 658244, 2021.
Article in English | MEDLINE | ID: mdl-33935654

ABSTRACT

Striatal dopamine transporters (DAT) powerfully regulate dopamine signaling, and can contribute risk to degeneration in Parkinson's disease (PD). DATs can interact with the neuronal protein α-synuclein, which is associated with the etiology and molecular pathology of idiopathic and familial PD. Here, we tested whether DAT function in governing dopamine (DA) uptake and release is modified in a human-α-synuclein-overexpressing (SNCA-OVX) transgenic mouse model of early PD. Using fast-scan cyclic voltammetry (FCV) in ex vivo acute striatal slices to detect DA release, and biochemical assays, we show that several aspects of DAT function are promoted in SNCA-OVX mice. Compared to background control α-synuclein-null mice (Snca-null), the SNCA-OVX mice have elevated DA uptake rates, and more pronounced effects of DAT inhibitors on evoked extracellular DA concentrations ([DA]o) and on short-term plasticity (STP) in DA release, indicating DATs play a greater role in limiting DA release and in driving STP. We found that DAT membrane levels and radioligand binding sites correlated with α-synuclein level. Furthermore, DAT function in Snca-null and SNCA-OVX mice could also be promoted by applying cholesterol, and using Tof-SIMS we found genotype-differences in striatal lipids, with lower striatal cholesterol in SNCA-OVX mice. An inhibitor of cholesterol efflux transporter ABCA1 or a cholesterol chelator in SNCA-OVX mice reduced the effects of DAT-inhibitors on evoked [DA]o. Together these data indicate that human α-synuclein in a mouse model of PD promotes striatal DAT function, in a manner supported by extracellular cholesterol, suggesting converging biology of α-synuclein and cholesterol that regulates DAT function and could impact DA function and PD pathophysiology.

2.
Eur J Pharmacol ; 883: 173377, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32687920

ABSTRACT

Lithium, commonly used to treat bipolar disorder, potentiates the ability of the muscarinic agonist pilocarpine to induce seizures in rodents. As this potentiation by lithium is reversed by the administration of myo-inositol, the potentiation may be mediated by inhibition of inositol monophosphatase (IMPase), a known target of lithium. Recently, we demonstrated that ebselen is a 'lithium mimetic' in regard to behaviours in both mice and man. Ebselen inhibits IMPase in vitro and lowers myo-inositol in vivo in the brains of mice and men, making ebselen the only known inhibitor of IMPase, other than lithium, that penetrates the blood-brain barrier. Our objective was to determine the effects of ebselen on sensitization to pilocarpine-induced seizures and neural activity. We administered ebselen at different doses and time intervals to mice, followed by injection of a sub-seizure dose of pilocarpine. We assessed seizure and neural activity by a subjective seizure rating scale, by monitoring tremors, and by induction of the immediate early gene c-fos. In contrast to lithium, ebselen did not potentiate the ability of pilocarpine to induce seizures. Unexpectedly, ebselen inhibited pilocarpine-induced tremor as well as pilocarpine-induced increases in c-fos mRNA levels. Both lithium and ebselen inhibit a common target, IMPase, but only lithium potentiates pilocarpine-induced seizures, consistent with their polypharmacology at diverse molecular targets. We conclude that ebselen does not potentiate pilocarpine-induced seizures and instead, reduces pilocarpine-mediated neural activation. This lack of potentiation of muscarinic sensitization may be one reason for the lack of side-effects observed with ebselen treatment clinically.


Subject(s)
Anticonvulsants/pharmacology , Azoles/pharmacology , Brain/drug effects , Lithium Chloride/toxicity , Neurons/drug effects , Organoselenium Compounds/pharmacology , Pilocarpine , Seizures/prevention & control , Animals , Anticonvulsants/toxicity , Azoles/toxicity , Brain/metabolism , Brain/physiopathology , CHO Cells , Calcium Signaling/drug effects , Cricetulus , Disease Models, Animal , Inositol Phosphates/metabolism , Isoindoles , Male , Mice , Neurons/metabolism , Organoselenium Compounds/toxicity , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/genetics , Receptors, Muscarinic/metabolism , Seizures/chemically induced , Seizures/metabolism , Seizures/physiopathology
3.
Neuropharmacology ; 63(6): 992-1001, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22828637

ABSTRACT

Studies of 5-HT-glutamate interactions suggest that activation of brain 5-HT(2A) receptors leads to an AMPA receptor-mediated induction of the immediate early (activity-dependent) gene, Arc (Arg3.1). In this respect, noradrenaline-glutamate interactions are poorly characterised. Here we investigated the influence on regional brain Arc gene expression of selective blockade of α(2)-adrenoceptors in rats. Several complementary techniques were used: qPCR (mRNA, discrete tissue punches), in situ hybridisation (mRNA, sections) and immunocytochemistry. The α(2)-adrenoceptor antagonist, RX 821002, dose-dependently and time-dependently (maximal effect 2 h) increased Arc mRNA levels as demonstrated both by qPCR and in situ hybridisation. The α(2)-adrenoceptor antagonist, atipamezole, also increased Arc mRNA in in situ hybridisation studies. Changes in Arc mRNA after RX 821002 were of similar magnitude in punches and intact tissue sections and region-specific, with effects being most pronounced in parietal cortex and caudate putamen, less robust in frontal cortex, and not detectable in hippocampal sub-regions. Both qPCR and in situ hybridisation studies demonstrated that RX 821002-induced Arc mRNA was blocked by the AMPA antagonist, GYKI 52466. Pretreatment with the NMDA antagonist MK 801 also prevented RX 821002-induced Arc mRNA, as did the mGluR5 antagonist MPEP, whilst the mGluR2/3 antagonist, LY341495, had no effect. Finally, immunocytochemical studies showed that RX 821002 increased Arc-immunoreactivity in cells in close apposition to α(2)-adrenoceptor-positive processes. Thus, employing three complementary techniques, these observations demonstrate that blockade of α(2)-adrenoceptors triggers brain expression of the immediate early gene, Arc, and that this effect involves the recruitment of AMPA, NMDA and mGluR5 but not mGluR2/3 glutamatergic receptors.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists/pharmacology , Brain Chemistry/drug effects , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Receptors, Glutamate/drug effects , Animals , Excitatory Amino Acid Antagonists/pharmacology , Idazoxan/analogs & derivatives , Idazoxan/pharmacology , Image Processing, Computer-Assisted , Imidazoles/pharmacology , Immunohistochemistry , In Situ Hybridization , Male , Polymerase Chain Reaction , RNA/biosynthesis , RNA/genetics , Rats , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5 , Receptors, AMPA/antagonists & inhibitors , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Serotonin/physiology
4.
J Pharmacol Exp Ther ; 340(3): 765-80, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22178753

ABSTRACT

The present studies characterized the functional profile of N-[4-methoxy-3-(4-methylpiperazin-1-yl)phenyl]-1,2-dihydro-3-H-benzo[e]indole-3-carboxamide) (S32212), a combined serotonin (5-HT)(2C) receptor inverse agonist and α(2)-adrenoceptor antagonist that also possesses 5-HT(2A) antagonist properties (J Pharmacol Exp Ther 340:750-764, 2012). Upon parenteral and/or oral administration, dose-dependent (0.63-40.0 mg/kg) actions were observed in diverse procedures. Both acute and subchronic administration of S32212 reduced immobility time in a forced-swim test in rats. Acutely, it also suppressed marble burying and aggressive behavior in mice. Long-term administration of S32212 was associated with rapid (1 week) and sustained (5 weeks) normalization of sucrose intake in rats exposed to chronic mild stress and with elevated levels of mRNA encoding brain-derived neurotrophic factor in hippocampus and amygdala (2 weeks). S32212 accelerated the firing rate of adrenergic perikarya in the locus coeruleus and elevated dialysis levels of noradrenaline in the frontal cortex and hippocampus of freely moving rats. S32212 also elevated the frontocortical levels of dopamine and acetylcholine, whereas 5-HT, amino acids, and histamine were unaffected. These neurochemical actions were paralleled by "promnemonic" properties: blockade of scopolamine-induced deficits in radial maze performance and social recognition and reversal of delay-induced impairments in social recognition, social novelty discrimination, and novel object recognition. It also showed anxiolytic actions in a Vogel conflict procedure. Furthermore, in an electroencephalographic study of sleep architecture, S32212 enhanced slow-wave and rapid eye movement sleep, while decreasing waking. Finally, chronic administration of S32212 neither elevated body weight nor perturbed sexual behavior in male rats. In conclusion, S32212 displays a functional profile consistent with improved mood and cognitive performance, together with satisfactory tolerance.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists/pharmacology , Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Brain Chemistry/drug effects , Indoles/pharmacology , Piperazines/pharmacology , Receptor, Serotonin, 5-HT2C/drug effects , Acetylcholine/analysis , Aggression/drug effects , Amygdala/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Dopamine/analysis , Dose-Response Relationship, Drug , Drug Inverse Agonism , Hippocampus/metabolism , Male , Mice , Motor Activity/drug effects , Norepinephrine/analysis , Rats , Rats, Wistar , Scopolamine/pharmacology , Sexual Behavior, Animal/drug effects , Sleep/drug effects , Sleep/physiology , Swimming
5.
J Psychopharmacol ; 26(4): 505-15, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22057017

ABSTRACT

Studies suggest that like selective 5-hydroxytryptamine (5-HT; serotonin) reuptake inhibitors, antagonists at neurokinin-1 receptors (NK(1)Rs) may have antidepressant and anxiolytic properties. NK(1)Rs are present in 5-HT innervated forebrain regions which may provide a common point of interaction between these two transmitter systems. This study aimed to investigate for cellular co-localization between NK(1)Rs and 5-HT receptor subtypes in mood-related brain regions in the rat forebrain. With experiments using fluorescence immunocytochemistry, double-labelling methods demonstrated a high degree of co-localization between NK(1)Rs and 5-HT(1A) receptors in most regions examined. Co-localization was highest in the medial septum (88% NK(1)R expressing cells were 5-HT(1A) receptor-positive) and hippocampal regions (e.g. dentate gyrus, 65%), followed by the lateral/basolateral amygdala (35%) and medial prefrontal cortex (31%). In contrast, co-localization between NK(1)Rs and 5-HT(2A) receptors was infrequent (< 8%) in most areas examined except for the hippocampus (e.g. CA3, 43%). Overall co-localization between NK(1)Rs and 5-HT(1A) receptors was much greater than that between NK(1)Rs and 5-HT(2A) receptors. Thus, these experiments demonstrate a high degree of co-localization between NK(1)Rs and 5-HT(1A) receptors in cortical and limbic regions of the rat forebrain. These findings suggest a novel site of interaction between NK(1)R antagonists and the 5-HT system.


Subject(s)
Prosencephalon/chemistry , Receptor, Serotonin, 5-HT1A/analysis , Receptor, Serotonin, 5-HT2A/analysis , Receptors, Neurokinin-1/analysis , Animals , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley
6.
Int J Neuropsychopharmacol ; 15(5): 617-29, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21733241

ABSTRACT

Evidence of early changes in neural plasticity may aid the prediction of rapid-onset antidepressant drugs. Here we compared the dual α2-adrenoceptor antagonist/5-HT-noradrenaline reuptake inhibitor (SNRI), S35966, to the SNRI, venlafaxine, with regards to their effect on rat brain expression of a panel of neural plasticity-related genes: Arc, BDNF, and VGLUT1, as well as Homer1a and Shank1B (not studied previously). Abundance of mRNA was determined by in-situ hybridization in cortical and hippocampal regions 2 h and 16 h following drug administration for 14, 7 and 1 d. After 14 d, both S35966 and venlafaxine increased mRNA of all genes, including Homer1a and Shank1B, and effects were similarly time- and region-dependent. After 7 d, S35966 elevated Arc, Shank1B and BDNF mRNA, whereas venlafaxine increased Shank1B mRNA only. Finally, after 1 d (acute administration), S35966 increased Arc and Homer1a mRNA whereas venlafaxine had no effect on any gene examined. In summary, a 14-d course of treatment with S35966 or venlafaxine induced similar region- and time-dependent increases in expression of neural plasticity-related genes including Shank1B and Homer1a. Some genes responded earlier to S35966, suggesting that drugs with combined α2-adrenoceptor antagonist/SNRI properties may elicit more rapid changes in markers of neural plasticity than a SNRI alone.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Cerebral Cortex/drug effects , Cyclohexanols/pharmacology , Gene Expression Regulation/drug effects , Hippocampus/drug effects , Nerve Tissue Proteins/drug effects , Neuronal Plasticity/drug effects , Animals , Antidepressive Agents, Second-Generation/administration & dosage , Cerebral Cortex/metabolism , Cyclohexanols/administration & dosage , Hippocampus/metabolism , Humans , Male , Nerve Tissue Proteins/genetics , Neuronal Plasticity/genetics , Norepinephrine/metabolism , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Time Factors , Venlafaxine Hydrochloride
7.
Psychopharmacology (Berl) ; 199(4): 549-68, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18523738

ABSTRACT

RATIONALE: Serotonin (5-HT)(2C) receptors are implicated in the control of mood, and their blockade is of potential interest for the management of anxiodepressive states. OBJECTIVES: Herein, we characterized the in vitro and in vivo pharmacological profile of the novel benzourea derivative, S32006. MATERIALS AND METHODS: Standard cellular, electrophysiological, neurochemical, and behavioral procedures were used. RESULTS: S32006 displayed high affinity for human (h)5-HT(2C) and h5-HT(2B) receptors (pK (i)s, 8.4 and 8.0, respectively). By contrast, it had negligible (100-fold lower) affinity for h5-HT(2A) receptors and all other sites examined. In measures of Gq-protein coupling/phospholipase C activation, S32006 displayed potent antagonist properties at h5-HT(2C) receptors (pK (B) values, 8.8/8.2) and h5-HT(2B) receptors (7.8/7.7). In vivo, S32006 dose-dependently (2.5-40.0 mg/kg, i.p. and p.o.) abolished the induction of penile erections and a discriminative stimulus by the 5-HT(2C) receptor agonist, Ro60,0175, in rats. It elevated dialysis levels of noradrenaline and dopamine in the frontal cortex of freely moving rats, and accelerated the firing rate of ventrotegmental dopaminergic and locus ceruleus adrenergic neurons. At similar doses, S32006 decreased immobility in a forced-swim test in rats, reduced the motor depression elicited by 5-HT(2C) and alpha(2)-adrenoceptor agonists, and inhibited both aggressive and marble-burying behavior in mice. Supporting antidepressant properties, chronic (2-5 weeks) administration of S32006 suppressed "anhedonia" in a chronic mild stress procedure and increased both expression of BDNF and cell proliferation in rat dentate gyrus. Finally, S32006 (0.63-40 mg/kg, i.p. and p.o) displayed anxiolytic properties in Vogel conflict and social interaction tests in rats. CONCLUSION: S32006 is a potent 5-HT(2C) receptor antagonist, and possesses antidepressant and anxiolytic properties in diverse rodent models.


Subject(s)
Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Indoles/pharmacology , Pyridines/pharmacology , Serotonin 5-HT2 Receptor Antagonists , Serotonin Antagonists/pharmacology , Aggression/drug effects , Animals , Binding, Competitive/drug effects , Biogenic Monoamines/physiology , Brain-Derived Neurotrophic Factor/metabolism , CHO Cells , Cell Proliferation/drug effects , Conflict, Psychological , Cricetinae , Cricetulus , Indoles/metabolism , Interpersonal Relations , Male , Mice , Motor Activity/drug effects , Penile Erection/drug effects , Pyridines/metabolism , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT2B/drug effects , Receptor, Serotonin, 5-HT2B/metabolism , Receptor, Serotonin, 5-HT2C/genetics , Receptor, Serotonin, 5-HT2C/metabolism , Recombinant Proteins/drug effects , Serotonin Antagonists/metabolism , Signal Transduction/drug effects , Swimming/psychology , Vocalization, Animal
8.
Brain Res ; 1101(1): 36-42, 2006 Jul 26.
Article in English | MEDLINE | ID: mdl-16790237

ABSTRACT

Nicotine has been shown to produce some beneficial effects in neurodegenerative disorders, and several studies have suggested that these effects may be mediated in part through the action of the neurotrophic factor BDNF. To further elucidate the interaction between nicotine and BDNF, we examined the effect of nicotine on the proliferation of the neuroblastoma cell line SH-SY5Y, which, following differentiation with retinoic acid, expresses both nicotinic receptors and the receptor for BDNF, TrkB. Both nicotine and the nicotinic alpha-7 selective agonist AR-17779 significantly increased cell proliferation albeit with bell-shaped dose-response kinetics. The blockade of this effect with either the alpha-7 nicotinic antagonist methyllycaconitine or the non-selective nicotinic antagonist mecamylamine indicated that the effect was mediated by nicotinic receptors. Prior addition of neutralising BDNF antibodies or of the tyrosine kinase inhibitor K252A (200 nM) completely blocked nicotine-induced proliferation, suggesting the involvement of TrkB signalling in the mediation of the effect. Nicotine also enhanced both the secretion of BDNF from the SH-SY5Y and cell surface density of TrkB receptors. These effects were abolished by pretreatment with MLA. These data indicate that activation of nicotinic receptors has effects upon the BDNF-TrkB pathway, inducing cell proliferation by promoting the release of BDNF, which in turn activates TrkB receptors.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cell Proliferation/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Aconitine/analogs & derivatives , Aconitine/pharmacology , Antibodies/pharmacology , Brain-Derived Neurotrophic Factor/immunology , Carbazoles/pharmacology , Carbon Isotopes/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Humans , Indole Alkaloids , Mecamylamine/pharmacology , Neuroblastoma/pathology , Nicotinic Antagonists/pharmacology , Receptor, trkB/metabolism , Thymidine/metabolism , Time Factors
9.
J Psychopharmacol ; 20(4): 570-6, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16204322

ABSTRACT

Antagonists at neurokinin 1 (NK1) receptors are attracting attention as potential treatments for depressive states in light of their actions in behavioural models predictive of antidepressant properties, their modulation of corticolimbic monoaminergic transmission, and their influence upon neural plasticity. Here, we evaluated the influence of NK1 receptor blockade upon two immediate early genes, Arc and c-fos, implicated in mechanisms of synaptic plasticity. Administration of the selective NK1 receptor antagonist, GR 205,171 (40, but not 1, 5 or 10 mg/kg i.p.), elicited a pronounced elevation in mRNA encoding Arc in both outer and inner layers of the parietal cortex of rat brain. This action was region-specific inasmuch as Arc expression did not change in other cortical territories examined including frontal cortex, nor in CA1, CA3 and the dentate gyrus of the hippocampus. In comparison to GR 205,171, its less active isomer GR 226,206 (1-40 mg/kg) did not significantly modify Arc gene expression in parietal cortex or other cortical areas. GR 205,171 (40 mg/kg) also increased the abundance of c-fos mRNA in outer and inner parietal cortex and caused a corresponding increase in c-fos immunoreactivity in this region. GR 226,206 (40 mg/kg i.p.) had no effect on either c-fos mRNA or protein in parietal cortex. In conclusion, administration of GR 205,171 elicits a stereospecific increase in Arc and c-fos expression in rat parietal cortex but not in other cortical regions. These data suggest that the parietal cortex plays a role in the central actions of NK1 receptor antagonists.


Subject(s)
Gene Expression/drug effects , Genes, Immediate-Early/drug effects , Neurokinin-1 Receptor Antagonists , Parietal Lobe/metabolism , Animals , Antiemetics/pharmacology , Cell Count , Genes, fos/drug effects , Genes, fos/genetics , Image Processing, Computer-Assisted , In Situ Hybridization , Male , Oncogene Proteins v-fos/biosynthesis , Oncogene Proteins v-fos/genetics , Parietal Lobe/drug effects , Piperidines/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Stereoisomerism , Tetrazoles/pharmacology
10.
J Pharmacol Exp Ther ; 309(3): 936-50, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14978196

ABSTRACT

In forced-swim tests in mice and rats, the novel D(3)/D(2) receptor agonist S32504 [(+)-trans-3,4,4a,5,6,10b-hexahydro-9-carbamoyl-4-propyl-2H-naphth[1,2-b]-1,4-oxazine] dose-dependently (0.04-2.5 mg/kg) and stereospecifically suppressed immobility compared with its enantiomer S32601 [(-)-trans-3,4,4a,5,6,10b-hexahydro-9-carbamoyl-4-propyl-2H-naphth-[1,2-b]-1,4-oxazine]. Ropinirole was less potent than S32504 in this procedure, and it was likewise less potent than S32504 (0.04-2.5 mg/kg) in attenuating motor-suppressant properties of the alpha(2)-adrenoceptor agonist S18616 [(S)-spiro[(1-oxa-2-amino-3-azacyclopent-2-ene)-4,2'-(1',2',3',4'-tetrahydronaphthalene)]]. In a learned helplessness paradigm, S32504 (0.08-2.5 mg/kg) suppressed escape failures. Furthermore, in a chronic mild stress model of anhedonia, S32504 (0.16-2.5 mg/kg) rapidly restored the suppression of sucrose consumption. S32504 inhibited marble-burying behavior in mice (0.04-0.16 mg/kg) and aggressive behavior in isolated mice (0.04-2.5 mg/kg): only higher doses of ropinirole mimicked these actions of S32504. In tests of anxiolytic activity, S32504 was more potent (0.0025-0.16 mg/kg) than ropinirole in suppressing fear-induced ultrasonic vocalizations, and S32601 was inactive. Furthermore, in contrast to ropinirole, S32504 modestly enhanced punished responses in a Vogel conflict procedure and increased open-arm entries in a plus-maze. At doses active in the above-described procedures, S32504 did not elicit hyperlocomotion. In the forced-swim, marble-burying, and ultrasonic vocalization models, actions of S32504 were blocked by the D(2)/D(3) antagonists haloperidol and raclopride and by the D(2) antagonist L741,626 [4-(4-chlorophenyl)-1-(1H-indol-3-ylmethyl)piperidin-4-ol], but not by the D(3) receptor antagonist S33084 [(3aR,9bS)-N-[4-(8-cyano-1,3a,4,9b-tetrahydro-3H-benzopyrano[3,4-c]pyrrole-2-yl)-butyl]-(4-phenyl)benzamide. Finally, chronic administration of S32504 did not, in contrast to venlafaxine, modify corticolimbic levels of serotonin(2A) receptors or brain-derived neurotrophic factor. In conclusion, S32504 displays a broad and distinctive profile of activity in models of potential antidepressive and anxiolytic properties. Its actions are more pronounced than those of ropinirole and principally involve engagement of D(2) receptors.


Subject(s)
Anti-Anxiety Agents/therapeutic use , Antidepressive Agents/therapeutic use , Dopamine Agonists/therapeutic use , Indoles/therapeutic use , Oxazines/therapeutic use , Aggression/drug effects , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Electrophysiology , Helplessness, Learned , Humans , Male , Maze Learning/drug effects , Mice , Motor Activity/drug effects , Neurochemistry , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT2A/metabolism , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3 , Sucrose/metabolism , Swimming , Vocalization, Animal/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...