Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Neuropsychopharmacol ; 14(9): 1165-78, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21087552

ABSTRACT

While dopamine D2 receptor partial agonists (PAs) have been long considered for treating schizophrenia, only one, aripiprazole, is clinically available for therapeutic use. This raises critically important questions as to what is unique about aripiprazole and to what extent animal models can predict therapeutic success. A number of PAs whose clinical fate is known: aripiprazole, preclamol, terguride, OPC-4392 and bifeprunox were compared to haloperidol (a reference antipsychotic) in several convergent preclinical animal models; i.e. amphetamine-induced locomotion (AIL) and conditioned avoidance response (CAR), predictive of antipsychotic effects; unilateral nigrostriatal lesioned rats, a model of hypo-dopaminergia; striatal Fos induction, a molecular marker for antipsychotic activity; and side-effects common to this class of drugs: catalepsy (motor side-effects) and prolactaemia. The results were compared across drugs with reference to their measured striatal D2 receptor occupancy. All the PAs occupied striatal D2 receptors in a dose dependent manner, inhibited AIL and CAR, and lacked motor side-effects or prolactinaemia despite D2 receptor occupancy exceeding 80%. At comparative doses, aripiprazole distinguished itself from the other PAs by causing the least rotation in the hypo-dopaminergic model (indicating the least intrinsic activity) and showed the highest Fos expression in the nucleus accumbens (indicating functional D2 antagonism). Although a number of PAs are active in antipsychotic animal models, not all of them succeed. Given that only aripiprazole is clinically available, it can be inferred that low functional intrinsic activity coupled with sufficient functional antagonism as reflected in the animal models may be a marker of success.


Subject(s)
Antipsychotic Agents/therapeutic use , Dopamine Agonists/therapeutic use , Receptors, Dopamine D2/agonists , Schizophrenia/drug therapy , Animals , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/adverse effects , Antipsychotic Agents/metabolism , Aripiprazole , Avoidance Learning/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dopamine Agonists/administration & dosage , Dopamine Agonists/adverse effects , Dopamine Agonists/metabolism , Dopamine Antagonists/administration & dosage , Dopamine Antagonists/adverse effects , Dopamine Antagonists/metabolism , Dopamine Antagonists/therapeutic use , Dopamine D2 Receptor Antagonists , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Immunohistochemistry , Locomotion/drug effects , Male , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Nucleus Accumbens/pathology , Piperazines/administration & dosage , Piperazines/adverse effects , Piperazines/metabolism , Piperazines/therapeutic use , Proto-Oncogene Proteins c-fos/metabolism , Quinolones/administration & dosage , Quinolones/adverse effects , Quinolones/metabolism , Quinolones/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D2/metabolism
2.
Psychopharmacology (Berl) ; 199(2): 275-89, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18521575

ABSTRACT

RATIONALE: l-Stepholidine, a dopamine D(2) antagonist with D(1) agonist activity, should in theory control psychosis and treat cognitive symptoms by enhancing cortical dopamine transmission. Though several articles describe its impact on the dopamine system, it has not been systematically evaluated and compared to available antipsychotics. MATERIALS AND METHODS: We examined its in vitro interaction with dopamine D(2) and D(1) receptors and compared its in vivo pharmacokinetic profile to haloperidol (typical) and clozapine (atypical) in animal models predictive of antipsychotic activity. RESULTS: In vitro, l-stepholidine showed significant activity on dopamine receptors, and in vivo, l-stepholidine demonstrated a dose-dependent striatal receptor occupancy (RO) at D(1) and D(2) receptors (D(1) 9-77%, 0.3-30 mg/kg; D(2) 44-94%, 1-30 mg/kg), though it showed a rather rapid decline of D(2) occupancy related to its quick elimination. In tests of antipsychotic efficacy, it was effective in reducing amphetamine- and phencyclidine-induced locomotion as well as conditioned avoidance response, whereas catalepsy and prolactin elevation, the main side effects, appeared only at high D(2)RO (>80%). This preferential therapeutic profile was supported by a preferential immediate early gene (Fos) induction in the nucleus accumbens over dorsolateral striatum. We confirmed its D(1) agonism in vitro, and then using D(2) receptor, knockout mice showed that l-stepholidine shows D(1) agonism in the therapeutic dose range. CONCLUSIONS: Thus, l-stepholidine shows efficacy like an "atypical" antipsychotic in traditional animal models predictive of antipsychotic activity and shows in vitro and in vivo D(1) agonism, and, if its rapid elimination does not limit its actions, it could provide a unique therapeutic approach to schizophrenia.


Subject(s)
Antipsychotic Agents/pharmacology , Berberine/analogs & derivatives , Dopamine D2 Receptor Antagonists , Receptors, Dopamine D1/agonists , Amphetamine/pharmacology , Animals , Antipsychotic Agents/pharmacokinetics , Avoidance Learning/drug effects , Berberine/pharmacokinetics , Berberine/pharmacology , Brain Chemistry/drug effects , Catalepsy/chemically induced , Catalepsy/psychology , Central Nervous System Stimulants/pharmacology , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Genes, fos/drug effects , Hallucinogens/pharmacology , Immunohistochemistry , Male , Mice , Mice, Knockout , Motor Activity/drug effects , Phencyclidine/pharmacology , Prolactin/blood , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/genetics , Receptors, Dopamine D2/genetics
3.
Neuropsychopharmacology ; 32(7): 1540-9, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17164815

ABSTRACT

There is growing interest in N-desmethylclozapine (NDMC), the major metabolite of clozapine, as a unique antipsychotic because it acts in vitro as a 5-HT(2) antagonist and as a partial agonist to dopamine D(2) and muscarinic receptors. To explore this, we compared NDMC to a typical (haloperidol), atypical (clozapine), and partial-agonist atypical (aripiprazole) antipsychotic in preclinical models. The comparison was carried out using: brain D(2) and 5-HT(2) receptor occupancy; animal models predictive of antipsychotic efficacy (amphetamine-induced hyperlocomotion (AIL) and conditioned avoidance response (CAR) models); measures predictive of side effects (catalepsy and prolactin elevation); and molecular markers predictive of antipsychotic action (striatal Fos induction). NDMC (10-60 mg/kg/s.c.) showed high 5-HT(2) (64-79%), but minimal D(2) occupancy (<15% at 60 mg/kg) 1 h after administration. In contrast to other antipsychotics, NDMC was not very effective in reducing AIL or CAR and showed minimal induction of Fos in the nucleus accumbens. However, like atypical antipsychotics, it showed no catalepsy, prolactin elevation, and minimal Fos in the dorsolateral striatum. It seems unlikely that NDMC would show efficacy as a stand-alone antipsychotic, however, its freedom from catalepsy and prolactin elevation, and its unique pharmacological profile (muscarinic agonism) may make it feasible to use this drug as an adjunctive treatment to existing antipsychotic regimens.


Subject(s)
Antipsychotic Agents/pharmacology , Brain Chemistry/drug effects , Brain/drug effects , Clozapine/analogs & derivatives , Psychotic Disorders/drug therapy , Animals , Aripiprazole , Avoidance Learning/drug effects , Avoidance Learning/physiology , Biomarkers/blood , Brain/metabolism , Brain/physiopathology , Brain Chemistry/physiology , Catalepsy/chemically induced , Clozapine/pharmacology , Dopamine Agonists/pharmacology , Drug Evaluation, Preclinical , Haloperidol/pharmacology , Male , Piperazines/pharmacology , Prolactin/blood , Proto-Oncogene Proteins c-fos/metabolism , Psychotic Disorders/metabolism , Psychotic Disorders/physiopathology , Quinolones/pharmacology , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D2/drug effects , Receptors, Dopamine D2/metabolism , Receptors, Serotonin, 5-HT2/drug effects , Receptors, Serotonin, 5-HT2/metabolism , Serotonin Antagonists/pharmacology , Treatment Outcome
4.
J Pharmacol Exp Ther ; 318(2): 810-8, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16648369

ABSTRACT

"Dopamine stabilizers" are a new class of compounds that have the ability to reverse both hypo- as well as hyperdopaminergia in vivo. This class, exemplified by the phenylpiperidines (S)-(-)-3-(3-methanesulfonyl-phenyl)-1-propyl-piperidine [(-)-OSU6162] and 4-(3-methanesulfonyl-phenyl)-1-propyl)-piperidine [ACR16] although lacking high in vitro binding affinity for dopamine D2 receptor [(-)-OSU6162, Ki = 447 nM; ACR16, Ki > 1 microM], shows functional actions, suggestive of their interaction. Hence, we evaluated in vivo D2 occupancy of these agents in rats and correlated it to observed effects in a series of behavioral, neurochemical, and endocrine models relevant to the dopamine system and antipsychotic effect. Both (-)-OSU6162 and ACR16 showed robust dose-dependent striatal D2 occupancy with ED50 values of 5.27 and 18.99 mg/kg s.c., respectively, and functional assays showed no partial agonism. Over an occupancy range of 37 to 87% (3-60 mg/kg) for (-)-OSU6162 and 35 to 74% (10-60 mg/kg) for ACR16, we observed both inhibitory (amphetamine-induced locomotor activity) and stimulatory effects (in habituated rats). Haloperidol, over a similar occupancy range (33-78%), potently inhibited psychostimulant activity and induced catalepsy, but it failed to activate habituated animals. In the conditioned avoidance response assay, ACR16 was clearly more efficacious than (-)-OSU6162. In addition, both these compounds demonstrated significant preferential Fos induction in the nucleus accumbens compared with the dorsolateral striatum, a strong predictor of atypical antipsychotic efficacy. The results suggest that dopamine stabilizers exhibit locomotor stabilizing as well as antipsychotic-like effects, with low motor side effect liability, in a dose range that corresponds to high D2 in vivo occupancy.


Subject(s)
Antipsychotic Agents , Dopamine Agents/pharmacology , Dyskinesia, Drug-Induced/psychology , Piperidines/pharmacology , Receptors, Dopamine D2/metabolism , Animals , Avoidance Learning/drug effects , Binding, Competitive/drug effects , Dihydroxyphenylalanine/metabolism , Dopamine Agents/metabolism , Dopamine Agents/toxicity , Dopamine Antagonists/metabolism , Haloperidol/metabolism , Immunohistochemistry , Male , Motor Activity/drug effects , Neostriatum/drug effects , Neostriatum/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Piperidines/metabolism , Piperidines/toxicity , Prolactin/blood , Proto-Oncogene Proteins c-fos/biosynthesis , Rats , Rats, Sprague-Dawley , Reserpine/metabolism
5.
Brain Res Mol Brain Res ; 136(1-2): 148-57, 2005 May 20.
Article in English | MEDLINE | ID: mdl-15893599

ABSTRACT

Orexins (hypocretins) have been implicated in the regulation of the normal sleep-wake cycle, in sensorimotor programming, and in other homeostatic and neuroregulatory processes. The present study examined the effects of sleep deprivation (SD) and sleep recovery on the expression of orexin 1 receptors (OX1R) and orexin 2 receptors (OX2R) throughout the brain. Rats were sacrificed either immediately after 96 h of sleep deprivation (SD group) or after SD followed by 24 h of sleep recovery (Rebound group). Prepro-orexin mRNA showed a non-significant increase in the SD group relative to controls, but a pronounced and significant increase in the Rebound group (+88%, P < 0.007). Similarly, sleep deprivation produced no effect on OX1R or OX2R mRNA levels. However, in the Rebound group, OX1R mRNA levels increased significantly, compared to either control or SD groups, in 37 of 92 brain regions analyzed, with particularly strong effects in the amygdala and hypothalamus. Changes in OX2R mRNA levels were also seen only in the sleep Rebound group, but they were fewer in number (10 out of 86 regions), were in the direction of decreased rather than increased expression, and were predominantly confined to cerebral cortical areas. These observations indicate that some factor associated with sleep recovery, possibly the compensatory increase in REM sleep, has strong effects on the orexin system at the mRNA level. They further indicate that,pOX1 and OX2 receptors are affected in opposite way and that the former are more vulnerable to these effects than the latter.


Subject(s)
Brain/metabolism , Gene Expression/physiology , Receptors, Neuropeptide/metabolism , Sleep/physiology , Animals , Brain/anatomy & histology , Gene Expression Regulation/physiology , In Situ Hybridization/methods , Intracellular Signaling Peptides and Proteins , Male , Neuropeptides/genetics , Neuropeptides/metabolism , Orexin Receptors , Orexins , Protein Precursors/genetics , Protein Precursors/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, G-Protein-Coupled , Receptors, Neuropeptide/genetics , Sleep Deprivation/metabolism , Time Factors
6.
Article in English | MEDLINE | ID: mdl-15694238

ABSTRACT

There is evidence to suggest that the antidepressant activity of sleep deprivation may be due to an enhancement of serotonergic and/or noradrenergic neurotransmission in brain. In the present study we examined the possibility that such changes may occur at the level of the norepinephrine (NET) and serotonin (SERT) and transporters. Rats were deprived of sleep for 96 h using the modified multiple platform method and then sacrificed for autoradiographic assessments of NET and SERT binding throughout the brain. [3H]Nisoxetine binding to the NE transporter was generally decreased in 44 of 45 areas examined, with significant reductions occurring in the anterior cingulate cortex (-16%), endopiriform n. (-18%), anterior olfactory n. (-19%), glomerular layer of olfactory bulb (-18%), ventral pallidum (-14%), medial preoptic area (-16%), retrochiasmatic/arcuate hypothalamus (-18%), anteromedial thalamic n. (-15%), and rostral raphe (-17%). In contrast, SERT binding measured with [11C]DASB showed no clear directional trends in 61 brain areas examined, but was significantly reduced in subdivisions of the anterior olfactory nucleus (-22%) and substantia nigra (-18%). Thus, sleep deprivation induced widespread decreases in NET binding, and fewer and well-localized decreases in SERT binding. Significant down-regulation in one brain region, the anterior olfactory nucleus, was observed in the case of both transporters. These results suggest that mechanisms involved in the antidepressant action of sleep deprivation may involve generalized NET down-regulation as well as decreased SERT binding in specific areas. Insofar as these changes may be associated with increased levels of serotonin (5-HT) and norepinephrine (NE) in the synapse, they suggest that sleep deprivation may share some basic mechanisms of action with several current antidepressant medications.


Subject(s)
Brain Chemistry/physiology , Brain/metabolism , Fluoxetine/analogs & derivatives , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/metabolism , Nerve Tissue Proteins/metabolism , Norepinephrine/metabolism , Protein Binding/physiology , Sleep Deprivation/metabolism , Animals , Autoradiography/methods , Benzylamines/pharmacology , Brain/anatomy & histology , Brain/drug effects , Carbon Isotopes/pharmacology , Fluoxetine/pharmacology , Male , Protein Binding/drug effects , Rats , Rats, Wistar , Serotonin Plasma Membrane Transport Proteins , Sleep Deprivation/physiopathology , Tritium/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...