Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 161
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Pharmacopsychiatry ; 49(2): 62-5, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26838118

ABSTRACT

INTRODUCTION: In schizophrenia early treatment may prevent disorder onset, or at least minimize its impact, suggesting possible neuroprotective properties of antipsychotics. The present study investigates the effects of chronic treatment with the atypical antipsychotic, risperidone, on locomotor sensitization in the subchronic phencyclidine-treated rat. METHODS: Rats were treated with phencyclidine sub-chronically (2 mg/kg bi-daily for one week followed by a one-week wash-out period) or vehicle. Half of the phencyclidine group was concurrently treated with risperidone (0.5 mg/kg IP) twice daily for 15 days, beginning 3 days before the start of phencyclidine administration. 6 weeks after treatment all rats were injected with a phencyclidine-challenge (3.2 mg/kg) and immediately after their locomotor activity measured for 20 min. RESULTS: Co-administration of risperidone at the time of phencyclidine administration significantly reduced the phencyclidine-challenge locomotor effect administered 6 weeks later. DISCUSSION: These results demonstrate that concurrent risperidone is neuroprotective, and clearly suggests its functionality can be translated to a clinical setting for treating the so-called prodrome.


Subject(s)
Central Nervous System Sensitization/drug effects , Motor Activity/drug effects , Neuroprotective Agents/pharmacology , Phencyclidine/administration & dosage , Phencyclidine/antagonists & inhibitors , Risperidone/administration & dosage , Risperidone/pharmacology , Animals , Male , Neuroprotective Agents/administration & dosage , Phencyclidine/pharmacology , Rats
2.
Chem Pharm Bull (Tokyo) ; 62(12): 1200-13, 2014.
Article in English | MEDLINE | ID: mdl-25450629

ABSTRACT

A novel class of phosphodiesterase 10A (PDE10A) inhibitors with improved metabolic stability in mouse liver microsomes were designed and synthesized starting from 2-({4-[1-methyl-4-(pyridin-4-yl)-1H-pyrazol-3-yl]phenoxy}methyl)quinoline (MP-10). Replacement of the phenoxymethyl part of MP-10 with an oxymethyl phenyl unit led to the identification of 2-[4-({[1-methyl-4-(pyridin-4-yl)-1H-pyrazol-3-yl]oxy}methyl)phenyl]quinoline (14), which showed moderate PDE10A inhibitory activity with improved metabolic stability in mouse and human liver microsomes over MP-10. Compound 14 showed high concentrations in plasma and brain after intraperitoneal administration and dose-dependently attenuated the hyperlocomotion induced by phencyclidine in mice, and oral administration of 14 (0.1, 0.3 mg/kg) also improved visual-recognition memory impairment in mice.


Subject(s)
Phosphodiesterase Inhibitors/chemical synthesis , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/drug effects , Quinolines/chemical synthesis , Quinolines/pharmacology , Animals , Brain/metabolism , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Humans , In Vitro Techniques , Injections, Intraperitoneal , Memory Disorders/chemically induced , Memory Disorders/drug therapy , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Models, Molecular , Motor Activity/drug effects , Phencyclidine/antagonists & inhibitors , Phencyclidine/pharmacology
3.
J Pharmacol Exp Ther ; 332(2): 622-31, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19864614

ABSTRACT

Subchronic administration of the N-methyl-d-aspartate receptor antagonist, phencyclidine (PCP), in rodents has been shown to produce impairment in novel object recognition (NOR), a model of visual learning and memory. We tested the hypothesis that the selective 5-HT(2A) inverse agonists, pimavanserin and (R)-(+)-alpha-(2,3-dimethoxyphenyl)-1-[2-(4-fluorophenylethyl]-4-piperidinemethanol (M100907), would potentiate subeffective doses of atypical antipsychotic drugs (APDs) to reverse the NOR deficits. Female rats received vehicle or PCP (2 mg/kg b.i.d.) for 7 days, followed by a 7-day washout. Pimavanserin (3 mg/kg) or M100907 (1 mg/kg) alone, or four atypicial APDs, risperidone (0.05-0.1 mg/kg), melperone (1-3 mg/kg), olanzapine (1-2 mg/kg), or N-desmethylclozapine (1-2 mg/kg), and the typical APD, haloperidol (0.05-0.1 mg/kg), were administered alone, or in combination with pimavanserin or M100907, before NOR testing. The exploration times of objects during 3-min acquisition and retention trials, separated by a 1-min interval, were compared by analysis of variance. Vehicle-, but not PCP-treated, animals, explored the novel object significantly more than the familiar in the retention trial (p < 0.05-0.01). Pretreatment with the higher doses of the atypical APDs, but not pimavanserin, M100907, or haloperidol alone, reversed the effects of PCP. The effect of risperidone was blocked by haloperidol pretreatment. Coadministration of pimavanserin or M100907, with ineffective doses of the atypical APDs, but not haloperidol, also reversed the PCP-induced deficit in NOR. These results support the importance of 5-hydroxytryptamine(2A) receptor blockade relative to D(2) receptor blockade in the ability of atypicals to ameliorate the effect of subchronic PCP, a putative measure of cognitive dysfunction in schizophrenia.


Subject(s)
Antipsychotic Agents/pharmacology , Drug Inverse Agonism , Phencyclidine/antagonists & inhibitors , Piperidines/pharmacology , Recognition, Psychology/drug effects , Serotonin 5-HT2 Receptor Antagonists , Urea/analogs & derivatives , Animals , Dopamine D2 Receptor Antagonists , Drug Interactions , Female , Fluorobenzenes/pharmacology , Phencyclidine/pharmacology , Random Allocation , Rats , Rats, Long-Evans , Urea/pharmacology
4.
Pharmacol Res ; 61(5): 430-6, 2010 May.
Article in English | MEDLINE | ID: mdl-20045056

ABSTRACT

Neboglamine is a functional modulator of the glycine site on the N-methyl-d-aspartate (NMDA) receptor. Dysfunction of this receptor has been associated with negative and cognitive symptoms in schizophrenia. Thus, we tested the hypothesis that neboglamine behaves as a potential antipsychotic. We compared the effects of neboglamine, D-serine, clozapine, and haloperidol on the expression of Fos-like immunoreactivity (FLI), a marker of neuronal activation, in rat forebrain. We also studied the effects of these agents on phencyclidine (PCP)-induced behaviour in rats, a model predictive of potential antipsychotic activity. Neboglamine, like haloperidol and clozapine, significantly increased the number of FLI-positive cells in the prefrontal cortex, nucleus accumbens, and lateral septal nucleus (3.2-, 4.8-, and 4.5-fold over control, respectively). Haloperidol dramatically increased FLI (390-fold over control) in the dorsolateral striatum, a brain region in which neboglamine and clozapine had no effect. The pattern of FLI induced by neboglamine closely matched that of d-serine, an endogenous agonist at the glycine site of NMDA receptors. Consistent with this finding, neboglamine restored NMDA-mediated neurotransmitter release in frontal cortex punches exposed to the NMDA antagonist PCP. In the behavioural model, all test compounds significantly inhibited PCP-induced hyperlocomotion. Unlike haloperidol and clozapine, neither neboglamine nor D-serine affected the basal levels of locomotor activity. Moreover, oral neboglamine dose-dependently inhibited both the hyperlocomotion and the frequency of rearing behaviour induced by PCP. These results, while confirming that the NMDA glycine site is a feasible target for activating the frontostriatal system, support the clinical evaluation of neboglamine as a treatment for schizophrenia.


Subject(s)
Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Pentanoic Acids/pharmacology , Receptors, N-Methyl-D-Aspartate/drug effects , Animals , Clozapine/pharmacology , Dose-Response Relationship, Drug , Hallucinogens/antagonists & inhibitors , Hallucinogens/pharmacology , Haloperidol/pharmacology , Immunohistochemistry , Male , Motor Activity/drug effects , Phencyclidine/antagonists & inhibitors , Phencyclidine/pharmacology , Prosencephalon/drug effects , Prosencephalon/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Receptors, Glycine/drug effects , Serine/metabolism
5.
J Neurochem ; 109(4): 1017-30, 2009 May.
Article in English | MEDLINE | ID: mdl-19519774

ABSTRACT

Early postnatal blockade of NMDA receptors by phencyclidine (PCP) causes cortical apoptosis in animals. This is associated with the development of schizophrenia-like behaviors in rats later in life. Recent studies show that the mechanism involves a loss of neurotrophic support from the phosphoinositol-3 kinase/Akt pathway, which is normally maintained by synaptic NMDA receptor activation. Here we report that activation of dopamine D1 receptors (D1R) with dihydrexidine (DHX) prevents PCP-induced neurotoxicity in cortical neurons by enhancing the efficacy of NMDAergic synapses. DHX increases serine phosphorylation of the NR1 subunit through protein kinase A activation and tyrosine phosphorylation of the NR2B subunit via Src kinase. DHX enhances recruitment of NR1 and NR2B, but not NR2A, into synapses. DHX also facilitated the synaptic response in cortical slices and this was blocked by an NR2B antagonist. DHX pre-treatment of rat pups prior to PCP on postnatal days 7, 9 and 11 inhibited PCP-induced caspase-3 activation on PN11 and deficits in pre-pulse inhibition of acoustic startle measured on PN 26-28. In summary, these data demonstrate that PCP-induced deficits in NMDA receptor function, neurotoxicity and subsequent behavioral deficits may be prevented by D1R activation in the cortex and further, it is suggested that D1R activation may be beneficial in treating schizophrenia.


Subject(s)
Dopamine Agonists/pharmacology , Excitatory Amino Acid Antagonists/toxicity , Membrane Proteins , Neuroprotective Agents , Neurotoxicity Syndromes/prevention & control , Phenanthridines/pharmacology , Phencyclidine/antagonists & inhibitors , Phencyclidine/toxicity , Receptors, Dopamine D1/agonists , Receptors, N-Methyl-D-Aspartate/drug effects , Synapses/drug effects , Animals , Cell Survival/drug effects , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/physiology , Electrophysiology , Enzyme-Linked Immunosorbent Assay , Female , Immunoprecipitation , Male , Membrane Proteins/metabolism , Patch-Clamp Techniques , Phosphorylation , Rats , Rats, Sprague-Dawley , Reflex, Startle/drug effects , Signal Transduction/drug effects , Tyrosine/metabolism
6.
Science ; 217(4566): 1272-3, 1982 Sep 24.
Article in English | MEDLINE | ID: mdl-6287581

ABSTRACT

Phencyclidine elicits hyperthermia at low doses and hypothermia at high doses in rats. Naloxone antagonizes both effects. Phencyclidine's effects on thermo-regulation are probably mediated by an interaction with a mu opiate receptor.


Subject(s)
Body Temperature Regulation/drug effects , Phencyclidine/pharmacology , Receptors, Opioid/drug effects , Animals , Dose-Response Relationship, Drug , Female , Naloxone/pharmacology , Phencyclidine/antagonists & inhibitors , Rats
7.
Neurotoxicology ; 30(1): 151-4, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19038286

ABSTRACT

Glutamate activation of the NMDA receptor is essential for neuronal differentiation, migration, and survival. Treatment with NMDA receptor antagonists, such as ketamine (KET) or phencyclidine (PCP), can trigger apoptosis in neonatal rats. However, L-carnitine (LC) treatment appears to prevent glutamate-induced toxicity in the developing CNS. Previously, we described altered preweaning behaviors (i.e., abnormal home cage, slant board and forelimb hang behaviors) resulting from neonatal PCP and KET treatment. Those adverse effects of KET were somewhat ameliorated by LC [Boctor SY, Wang C, Ferguson SA. Neonatal PCP is more potent than ketamine at modifying preweaning behaviors of Sprague-Dawley rats. Toxicol Sci 2008;106:172-9]. Here, a portion of those subjects were evaluated for prepulse inhibition (PPI) of the acoustic startle response at postnatal day (PND) 25 since previous reports described PCP-induced effects on this response. Rats were subcutaneously treated with: saline; 10 mg/kg PCP (1x/day) on PNDs 7, 9 and 11; 20 mg/kg KET (6 injections every 2h on PND 7); or a similar regimen of ketamine and 250 mg/kg LC on PND 7, with a single injection of 250 mg/kg LC on PNDs 8-11 (KLC). Male and female rats were assessed using a standard PPI paradigm with prepulses of 68, 78 and 82 dB. Body weight was decreased 17-21% and whole brain weight was decreased 10% in PCP-treated rats. Specifically, cerebellar weight was significantly less in PCP-treated rats relative to control. Despite the magnitude of those PCP-induced changes, startle response in normal pulse only trials and percent of PPI in PCP-, KET-, and KLC-treated groups were comparable to controls. Average latency to maximum startle was 2.6 ms less in females than males (p<0.007); there were no other significant sex effects. The lack of neonatal PCP treatment on later PPI is similar to that reported by Rasmussen et al. [Rasmussen BA, O'Neil J, Manaye KF, Perry DC, Tizabi Y. Long-term effects of developmental PCP administration on sensorimotor gating in male and female rats. Psychopharmacology (Berl) 2007; 190: 43-9.], and indicates that neonatal PCP-induced effects on PPI [Wang C, McInnis J, Ross-Sanchez M, Shinnick-Gallagher P, Wiley JL, Johnson KM. Long-term behavioral and neurodegenerative effects of perinatal phencyclidine administration: implications for schizophrenia. Neuroscience 2001; 107: 535-50.] appear difficult to replicate.


Subject(s)
Inhibition, Psychological , Ketamine/administration & dosage , Phencyclidine/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Animals, Newborn , Body Weight/drug effects , Carnitine/pharmacology , Cerebellum/drug effects , Female , Ketamine/antagonists & inhibitors , Ketamine/pharmacology , Male , Organ Size/drug effects , Phencyclidine/administration & dosage , Phencyclidine/antagonists & inhibitors , Postpartum Period , Rats , Rats, Sprague-Dawley , Reflex, Startle/drug effects , Sex Characteristics
8.
Georgian Med News ; (169): 81-7, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19430052

ABSTRACT

Finding about structural and functional relation between NMDA receptors specific binding and phencyclidine sites was very important for a possible modulation of NMDA receptors' function. We have therefore got interested what would happen with EEG and vegetative patterns of PS in the case when NMDA receptors function is modulated by blocking of phencyclidines' site. Consequently, we studied the effects of Trihexyphenydil, the structural analog of phencyclidine, on neocortical and hippocampal electrical activity in SWC. On cats (n=5) metallic electrodes were implanted under Nembutal anesthesia. EEG registration lasting 12 hr daily started after animals' recovery. Trihexyphenydil was administered intraperitoneally (0.5 mg/kg - 1 mg/kg). Statistical processing was made by Students' t-test. Trihexyphenydil resulted in dissociated triggering of PS. Rapid eye movements and PGO waves appeared on the face of active waking state. Therefore on the background of behavioral active waking according to electrical activity of the visual cortex and rapid eye movements, electrographic patterns of paradoxical sleep were recorded. Thus in our experiments it was shown firstly that the mechanism of hallucinogenic action of Trihexyphenydil is closely related to the disturbance of paradoxical sleep integrity. Blocking of NMDA receptors phencyclidines site and therefore functional modulation of these receptors produce the splitting of PS patterns and their intrusion in waking state. Such an effect never takes place in normal conditions since the waking system has the powerful inhibitory influence on the PS triggering system. Suggestion is make that NMDA glutamate receptors must be involved in mechanisms providing structural and functional integrity of PS and that fulfillment of such function is possible in the case when the NMDA receptors phencyclidine site isn't in blocked state. Normal functioning of NMDA receptors phencyclidine site represents the mechanism which inhibits and/or hampers appearance of hallucination. NMDA glutamate receptors, possessing phencyclidine site, are implicated in the mechanisms providing structural and functional integrity of PS.


Subject(s)
Hippocampus/drug effects , Neocortex/drug effects , Phencyclidine/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/drug effects , Sleep/drug effects , Trihexyphenidyl/pharmacology , Wakefulness/drug effects , Animals , Cats , Circadian Rhythm/drug effects , Electroencephalography/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/physiology , Neocortex/physiology , Phencyclidine/chemistry , Phencyclidine/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Sleep/physiology , Trihexyphenidyl/chemistry , Wakefulness/physiology
9.
Neuropharmacology ; 157: 107675, 2019 10.
Article in English | MEDLINE | ID: mdl-31233824

ABSTRACT

Non-coding RNA molecules such as miRNAs have emerged as critical regulators of neuronal functions. The present study investigates a role for miRNA-143, a highly conserved miRNA, in locomotorhyperactivity induced bythe psychotomimetic phencyclidine (PCP), a non-selective antagonist of the N-methyl-d-aspartate (NMDA) glutamate receptor. Following acute PCP administration to mice, the content of miRNA-143 was reduced in plasma, prefrontal cortex (PFC) and hippocampus, reaching a minimum after 2 h. The antipsychotics haloperidol and clozapine attenuated hyperlocomotion and the decrease in miR-143 expression induced by PCP, as did the selective D2 dopamine receptor antagonist eticlopride but not the selective D1 antagonist SCH23390. To further confirm D2 receptor-mediated miRNA-143 expression, HT-22 neuronal cell line and primary cortical cultured neuronswere studied. Stimulation of D2 receptors with the selective D2 receptor agonist quinpirole decreased expression of miRNA-143 in a time-dependent manner. This inhibition was blocked by pretreatment with eticlopride, indicating that the D2 receptor directly regulates the expression of miRNA-143. We further demonstrated that miRNA-143 directly targeted to the 3' un-translated region of neuregulin-1 (NRG1) mRNA to reduce protein expression of NRG1 in HT-22 cells and that administration of the D2 receptor agonist quinpirole to mice enhanced expression of NRG1 in PFC. The present data provide the first evidence that D2 receptors are involved in the expression ofmiRNA-143 in association with antipsychotic drug action and the developmental regulator NRG1.


Subject(s)
MicroRNAs/biosynthesis , Neuregulin-1/biosynthesis , Phencyclidine/pharmacology , Receptors, Dopamine D2/physiology , Animals , Benzazepines/pharmacology , Clozapine/pharmacology , Dopamine D2 Receptor Antagonists/pharmacology , Haloperidol/pharmacology , Hippocampus/metabolism , Male , Mice , Mice, Transgenic , MicroRNAs/blood , MicroRNAs/drug effects , Motor Activity/drug effects , Phencyclidine/antagonists & inhibitors , Prefrontal Cortex/metabolism , Primary Cell Culture , Quinpirole/pharmacology , Receptors, Dopamine D2/agonists , Salicylamides/pharmacology
10.
Psychopharmacology (Berl) ; 197(3): 457-64, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18251012

ABSTRACT

RATIONALE: Neurodegenerative changes have been suggested to provide a basis for the pathophysiology of schizophrenia. T-817MA (1-{3-[2-(1-benzothiophen-5-yl) ethoxy] propyl} azetidin-3-ol maleate) is a novel compound with neuroprotective and neurite-outgrowth effects, as elicited in rat primary cultured neurons. OBJECTIVES: We examined the effect of T-817MA on phencyclidine (PCP)-induced disruption of prepulse inhibition (PPI), a measure of sensorimotor gating, in male Wistar rats. MATERIALS AND METHODS: In chronic experiments, male Wistar rats were injected intermittently with PCP (2.0 mg/kg, i.p., three times per week) or vehicle (saline, 2.0 ml/kg) for 1 month. T-817MA (0.21 or 0.07 mg/ml, p.o.) or distilled water was administered throughout the study period. In an acute experiment, T-817MA (8.4 mg/kg, p.o.) or distilled water was administered, followed by treatment with PCP (2.0 mg/kg, i.p.) or vehicle (saline, 2.0 ml/kg), before PPI measurements. RESULTS: Intermittent administration of PCP for 1 month induced persistent disruption of PPI. Coadministration of T-817MA at 0.21 mg/ml but not 0.07 mg/ml completely blocked PCP-induced disruption of PPI, whereas T-817MA (0.21 mg/ml) by itself did not show a significant effect on PPI in control rats. On the other hand, single administration of T-817MA did not affect PPI disruption by acute treatment with PCP. CONCLUSIONS: These results suggest that T-817MA is effective in ameliorating sensorimotor gating deficits caused by chronic PCP treatment, possibly via neuroprotective actions. Our findings provide a novel therapeutic approach for patients with schizophrenia.


Subject(s)
Attention/drug effects , Excitatory Amino Acid Antagonists/toxicity , Inhibition, Psychological , Maleates/pharmacology , Neuroprotective Agents/pharmacology , Phencyclidine/antagonists & inhibitors , Phencyclidine/toxicity , Reflex, Startle/drug effects , Thiophenes/pharmacology , Acoustic Stimulation , Animals , Dose-Response Relationship, Drug , Drug Administration Schedule , Injections, Intraperitoneal , Male , Rats , Rats, Wistar
11.
Psychopharmacology (Berl) ; 201(2): 273-84, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18704372

ABSTRACT

RATIONALE: Increasing experimental evidence suggests that impaired N-methyl-D: -aspartic acid (NMDA) receptor (NMDAr) function could be a key pathophysiological determinant of schizophrenia. Agonists at the allosteric glycine (Gly) binding site of the NMDA complex can promote NMDAr activity, a strategy that could provide therapeutic efficacy for the disorder. NMDAr antagonists like phencyclidine (PCP) can induce psychotic and dissociative symptoms similar to those observed in schizophrenia and are therefore widely used experimentally to impair NMDA neurotransmission in vivo. OBJECTIVES: In the present study, we used pharmacological magnetic resonance imaging (phMRI) to investigate the modulatory effects of endogenous and exogenous agonists at the NMDAr Gly site on the spatiotemporal patterns of brain activation induced by acute PCP challenge in the rat. The drugs investigated were D: -serine, an endogenous agonist of the NMDAr Gly site, and SSR504734, a potent Gly transporter type 1 (GlyT-1) inhibitor that can potentiate NMDAr function by increasing synaptic levels of Gly. RESULTS: Acute administration of PCP induced robust and sustained activation of discrete cortico-limbo-thalamic circuits. Pretreatment with D: -serine (1 g/kg) or SSR504734 (10 mg/kg) completely inhibited PCP-induced functional activation. This effect was accompanied by weak but sustained deactivation particularly in cortical areas. CONCLUSIONS: These findings suggest that agents that stimulate NMDAr via Gly co-agonist site can potentiate NMDAr activity in the living brain and corroborate the potential for this class of drugs to provide selective enhancement of NMDAr neurotransmission in schizophrenia.


Subject(s)
Glycine/metabolism , Glycine/pharmacology , Magnetic Resonance Imaging , Phencyclidine/adverse effects , Receptors, N-Methyl-D-Aspartate/agonists , Animals , Benzamides/pharmacology , Binding Sites , Blood Volume/drug effects , Brain/anatomy & histology , Brain/blood supply , Brain/drug effects , Dose-Response Relationship, Drug , Glycine Plasma Membrane Transport Proteins/antagonists & inhibitors , Injections, Intravenous , Male , Models, Theoretical , Phencyclidine/antagonists & inhibitors , Piperidines/pharmacology , Rats , Rats, Sprague-Dawley , Serine/pharmacology , Time Factors
12.
Schizophr Res ; 105(1-3): 224-35, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18710798

ABSTRACT

INTRODUCTION: Amisulpride's high and selective affinity for dopamine D2/3 (Ki 1.3/2.4 nM) receptors, lack of affinity for serotonin receptors, and its unusually high therapeutic doses (400-800 mg/day) makes it unique among atypical antipsychotics and prompted us to compare its actions with other antipsychotics in animal models. METHODS: Amisulpride's effects on amphetamine and phencyclidine induced locomotor activity (AIL/PIL), conditioned avoidance response, catalepsy (CAT), subcortical Fos expression, and plasma prolactin was correlated to its time-course striatal D2/3 and prefrontal 5-HT2 receptor occupancy (D(2/3)/5-HT2RO); in comparison to haloperidol, clozapine, and risperidone. RESULTS: Unlike the atypicals clozapine and risperidone, amisulpride lacked 5-HT2RO and showed a 'delayed' pattern of D2/3RO: 43, 60 and 88% after 1, 2 and 6 h (100 mg/kg), respectively, despite a quick onset (1 h) and decline (6 h) of prolactin elevation. While haloperidol and risperidone were effective at D2RO>60%, clozapine at D2/3RO<50%, amisulpride was effective only when its D2RO exceeded 60% with a delayed latency and lasted longer than other antipsychotics. CAT was observed for haloperidol and risperidone when D2RO exceeded 80%, while in the case of amisulpride, CAT was not observed even when doses exceeded 90% D2/3RO. Amisulpride also displayed functional limbic selectivity in Fos expression like the other atypicals. CONCLUSIONS: Amisulpride's "delayed" functional profile on acute administration and the need for high doses is most likely due to its poor blood-brain-barrier penetration; however, it is distinct from other atypicals in showing low motor side-effects, activity against phencyclidine, and a mesolimbic preference, despite no action on serotonin receptors.


Subject(s)
Antipsychotic Agents/pharmacology , Brain/drug effects , Receptors, Dopamine/drug effects , Receptors, Serotonin/drug effects , Sulpiride/analogs & derivatives , Amisulpride , Amphetamine/antagonists & inhibitors , Amphetamine/pharmacology , Animals , Antipsychotic Agents/metabolism , Basal Ganglia/drug effects , Basal Ganglia/metabolism , Behavior, Animal/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/metabolism , Clozapine/metabolism , Clozapine/pharmacology , Conditioning, Psychological/drug effects , Haloperidol/metabolism , Haloperidol/pharmacology , Male , Models, Animal , Motor Activity/drug effects , Phencyclidine/antagonists & inhibitors , Phencyclidine/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Prolactin/blood , Prolactin/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Dopamine/metabolism , Receptors, Dopamine D2/drug effects , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/drug effects , Receptors, Dopamine D3/metabolism , Receptors, Serotonin/metabolism , Risperidone/metabolism , Risperidone/pharmacology , Sulpiride/metabolism , Sulpiride/pharmacology
13.
Neurotox Res ; 14(2-3): 129-40, 2008 Oct.
Article in English | MEDLINE | ID: mdl-19073421

ABSTRACT

Cognitive deficits in schizophrenia are associated with prefrontal cortex (PFC) abnormalities. Schizophrenic patients show a reduced performance in tasks engaging the PFC and a reduction of markers of cellular integrity and function. Non-competitive N-methyl-D-aspartate (NMDA) receptor antagonists are widely used as pharmacological models of schizophrenia due to their ability to exacerbate schizophrenia symptoms in patients and to elicit psychotomimetic actions in healthy volunteers. Also, these drugs evoke behavioral alterations in experimental animals that resemble schizophrenia symptoms. The PFC seems to be a key target area for these agents. However, the cellular and network elements involved are poorly known. Cognitive deficits are of particular interest since an early antipsychotic-induced improvement in cognitive performance predicts a better long-term clinical outcome. Here we report that the non-competitive NMDA receptor antagonist phencyclidine (PCP) induces a marked disruption of the activity of PFC. PCP administration increased the activity of a substantial proportion of pyramidal neurons, as evidenced by an increase in discharge rate and in c-fos expression. Examination of the effects of PCP on other brain areas revealed an increased c-fos expression in a number of cortical and subcortical areas, but notably in thalamic nuclei projecting to the PFC. The administration of classical (haloperidol) and/or atypical (clozapine) antipsychotic drugs reversed PCP effects. These results indicate that PCP induces a marked disruption of the network activity in PFC and that antipsychotic drugs may partly exert their therapeutic effect by normalizing hyperactive cortico-thalamocortical circuits.


Subject(s)
Antipsychotic Agents/pharmacology , Phencyclidine/pharmacology , Prefrontal Cortex/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Thalamic Nuclei/drug effects , Animals , Cerebral Cortex/metabolism , Clozapine/pharmacology , Haloperidol/pharmacology , Male , Neural Pathways , Phencyclidine/antagonists & inhibitors , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiology , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Pyramidal Cells/physiology , Rats , Rats, Wistar , Somatosensory Cortex/metabolism , Thalamic Nuclei/metabolism
14.
Pharmacol Biochem Behav ; 88(3): 189-95, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17936884

ABSTRACT

It has been observed that agents with agonist activity at 5-HT2A receptors prevent neurotoxicity induced by the non-competitive NMDA antagonist, dizocilpine (MK-801). Subsequent behavioral studies reported complete antagonism by LSD and DOM of the stimulus effects of the related NMDA antagonist, phencyclidine [PCP]. The present study sought to extend those observations to include other psychoactive drugs. Male F-344 rats were trained in a 2-lever, fixed-ratio 10, food-reinforced task with PCP (3.0 mg/kg; IP; 30 min pretreatment) as a discriminative stimulus. Tests of generalization were then conducted using the training dose of PCP in combination with a range of doses of DOM, LSD, d-amphetamine, MDMA, psilocybin, buspirone, and GHB. All of the drugs tested in combination with PCP produced a statistically significant diminution of PCP-appropriate responding but for none was antagonism complete. These data, obtained using a stimulus control model of the hallucinogenic effects of PCP, fail to support the hypothesis that LSD and DOM completely antagonize stimulus control by PCP. Instead, the data suggest complex interactions between PCP-induced stimulus control and a variety of psychoactive drugs including GHB, an agent with no known affinity for serotonergic receptors.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Hallucinogens/antagonists & inhibitors , Hallucinogens/pharmacology , Phencyclidine/antagonists & inhibitors , Phencyclidine/pharmacology , Psychotropic Drugs/pharmacology , Anesthetics, Intravenous/pharmacology , Animals , Conditioning, Operant/drug effects , Dopamine Agents/pharmacology , Dose-Response Relationship, Drug , Male , Rats , Rats, Inbred F344 , Serotonin Agents/pharmacology , Sodium Oxybate/pharmacology
15.
Pharmacol Biochem Behav ; 90(4): 569-77, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18544461

ABSTRACT

We sought to determine the relationship between phencyclidine (PCP)-induced alterations in behavior and NMDAR expression in the cortex by examining the effect of anti-schizophrenic drug treatment on both. Sprague-Dawley rat pups were pretreated with risperidone or olanzapine prior to treatment with PCP on postnatal day 7 (PN7) or sub-chronically on PN7, 9, and 11. Pre-pulse inhibition (PPI) of acoustic startle was measured on PN24-26 and following a challenge dose of 4 mg/kg PCP, locomotor activity was measured on PN28-35. PCP treatment on PN7 did not cause a deficit in PPI, but did cause locomotor sensitization. This was prevented by both antipsychotics. PCP treatment on PN7 caused an up-regulation of NR1 and NR2B, which was not affected by either anti-schizophrenic drug. PCP treatment on PN7, 9, and 11 caused a deficit in PPI and a sensitized locomotor response to PCP challenge as well as an up-regulation of NR1 and NR2A, all of which were prevented by both atypical anti-schizophrenic drugs. These data support the hypothesis that sub-chronic, but not single injection PCP treatment in developing rats results in behavioral alterations that are sensitive to antipsychotic drugs and these behavioral changes observed could be related to up-regulation of cortical NR1/NR2A receptors.


Subject(s)
Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Cerebral Cortex/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Phencyclidine/antagonists & inhibitors , Phencyclidine/pharmacology , Receptors, N-Methyl-D-Aspartate/drug effects , Schizophrenia/drug therapy , Animals , Blotting, Western , Cerebral Cortex/drug effects , Down-Regulation/drug effects , Excitatory Amino Acid Antagonists/administration & dosage , Female , Male , Motor Activity/drug effects , Neostriatum/drug effects , Neostriatum/metabolism , Phencyclidine/administration & dosage , Rats , Rats, Sprague-Dawley , Reflex, Startle/drug effects , Subcellular Fractions/drug effects
16.
J Med Chem ; 61(6): 2303-2328, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29350927

ABSTRACT

Multiple therapeutic opportunities have been suggested for compounds capable of selective activation of metabotropic glutamate 3 (mGlu3) receptors, but small molecule tools are lacking. As part of our ongoing efforts to identify potent, selective, and systemically bioavailable agonists for mGlu2 and mGlu3 receptor subtypes, a series of C4ß-N-linked variants of (1 S,2 S,5 R,6 S)-2-amino-bicyclo[3.1.0]hexane-2,6-dicarboxylic acid 1 (LY354740) were prepared and evaluated for both mGlu2 and mGlu3 receptor binding affinity and functional cellular responses. From this investigation we identified (1 S,2 S,4 S,5 R,6 S)-2-amino-4-[(3-methoxybenzoyl)amino]bicyclo[3.1.0]hexane-2,6-dicarboxylic acid 8p (LY2794193), a molecule that demonstrates remarkable mGlu3 receptor selectivity. Crystallization of 8p with the amino terminal domain of hmGlu3 revealed critical binding interactions for this ligand with residues adjacent to the glutamate binding site, while pharmacokinetic assessment of 8p combined with its effect in an mGlu2 receptor-dependent behavioral model provides estimates for doses of this compound that would be expected to selectively engage and activate central mGlu3 receptors in vivo.


Subject(s)
Bridged Bicyclo Compounds/chemical synthesis , Bridged Bicyclo Compounds/pharmacology , Excitatory Amino Acid Agonists/chemical synthesis , Excitatory Amino Acid Agonists/pharmacology , Receptors, Metabotropic Glutamate/agonists , Animals , Bridged Bicyclo Compounds/pharmacokinetics , Crystallography, X-Ray , Cyclic AMP/pharmacology , Excitatory Amino Acid Agonists/pharmacokinetics , Excitatory Amino Acid Antagonists/pharmacology , Humans , Male , Models, Molecular , Molecular Docking Simulation , Molecular Structure , Motor Activity/drug effects , Neurons/drug effects , Neurons/metabolism , Phencyclidine/antagonists & inhibitors , Phencyclidine/pharmacology , Protein Binding , Rats , Rats, Sprague-Dawley
17.
J Neurosurg Anesthesiol ; 30(4): 359-367, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29076977

ABSTRACT

This study examines the direct effects of 3 noncompetitive N-methyl-D-aspartate receptor antagonists, phencyclidine (PCP), (+)MK-801, and (-)MK-801, on bovine middle cerebral arteries (BMCA). Rings of BMCA were mounted in isolated tissue chambers equipped with isometric tension transducers to obtain pharmacologic dose-response curves. In the absence of endogenous vasoconstrictors, the 3 N-methyl-D-aspartate antagonists each produced direct constriction of BMCA. The thromboxane A2 receptor antagonist SQ-29,548, the TxA2 synthase inhibitor furegrelate, the calcium antagonist nimodipine, and calcium-deficient media all inhibited maximal phencyclidine or (+)MK-801-induced constriction. Direct constriction by PCP or (+)MK-801 was independent of the presence of endothelium. When BMCA were preconstricted with potassium-depolarizing solution, PCP, (+)MK-801, and (-)MK-801 each produced only concentration-dependent relaxation. When BMCA were preconstricted with the stable TxA2 analog U-46,619 and exposed to increasing concentrations of PCP, (+)MK-801, or (-)MK-801, tension increased. Thromboxane A2 may contract BMCA by acting as a potassium channel blocker; iberiotoxin and tetraethylammonium both constrict BMCA. In Ca-deficient media containing either potassium or U-46,619, phencyclidine and (+)MK-801 each produced competitive inhibition of subsequent Ca-induced constriction. In additional experiments, arterial strips were mounted in isolated tissue chambers to directly measure calcium uptake, using Calcium as a radioactive tracer. Both phencyclidine and (+)MK-801 blocked potassium-stimulated or U-46,619-stimulated Ca uptake into arterial strips. These results suggest that phencyclidine and (+)MK-801 have 2 separate actions on BMCA. They may constrict arterial rings by releasing TxA2 from cerebrovascular smooth muscle, and relax arterial rings by acting as calcium antagonists.


Subject(s)
Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Middle Cerebral Artery/drug effects , Phencyclidine/pharmacology , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cattle , Dose-Response Relationship, Drug , In Vitro Techniques , Isometric Contraction/drug effects , Middle Cerebral Artery/metabolism , Muscle Relaxation/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Phencyclidine/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Thromboxane A2/pharmacology
18.
Neuropharmacology ; 140: 246-259, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30005976

ABSTRACT

Group II metabotropic glutamate receptors (mGluR2 and mGluR3) are implicated in a number of psychiatric disorders. They also control sleep-wake architecture and may offer novel therapeutic targets. However, the roles of the mGluR2 versus mGluR3 subtypes are not well understood. Here, we have taken advantage of the recently described mutant strain of Han Wistar rats, which do not express mGluR2 receptors, to investigate behavioural, sleep and EEG responses to mGluR2/3 ligands. The mGluR2/3 agonist, LY354740 (10 mg/kg), reversed amphetamine- and phencyclidine-induced locomotion and rearing behaviours in control Wistar but not in mGluR2 lacking Han Wistar rats. In control Wistar but not in Han Wistar rats the mGluR2/3 agonist LY379268 (3 & 10 mg/kg) induced REM sleep suppression with dose-dependent effects on wake and NREM sleep. By contrast, the mGluR2/3 antagonist LY3020371 (3 & 10 mg/kg) had wake-promoting effects in both rat strains, albeit smaller in the mGluR2-lacking Han Wistar rats, indicating both mGluR2 and mGluR3-mediated effects on wakefulness. LY3020371 enhanced wake cortical oscillations in the theta (4-9 Hz) and gamma (30-80 Hz) range in both Wistar and Han Wistar rat strains, whereas LY379268 reduced theta and gamma oscillations in control Wistar rats, with minimal effects in Han Wistar rats. Together these studies illustrate the significant contribution of mGluR2 to the antipsychotic-like, sleep and EEG effects of drugs acting on group II mGluRs. However, we also provide evidence of a role for mGluR3 activity in the control of sleep and wake cortical theta and gamma oscillations.


Subject(s)
Antipsychotic Agents/pharmacology , Receptors, Metabotropic Glutamate/physiology , Sleep/physiology , Wakefulness/drug effects , Wakefulness/physiology , Amino Acids/pharmacology , Amphetamine/antagonists & inhibitors , Amphetamine/pharmacology , Animals , Bridged Bicyclo Compounds/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cyclohexanes/pharmacology , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/pharmacology , Gamma Rhythm/drug effects , Gamma Rhythm/physiology , Locomotion/drug effects , Locomotion/physiology , Male , Motor Activity/drug effects , Motor Activity/physiology , Mutation , Phencyclidine/antagonists & inhibitors , Phencyclidine/pharmacology , Rats , Receptors, Metabotropic Glutamate/deficiency , Receptors, Metabotropic Glutamate/genetics , Sleep/drug effects , Theta Rhythm/drug effects , Theta Rhythm/physiology
19.
Psychopharmacology (Berl) ; 192(1): 9-15, 2007 May.
Article in English | MEDLINE | ID: mdl-17235609

ABSTRACT

RATIONALE: The cognitive and attentional deficits observed in schizophrenic patients are now considered central to the pathophysiology of the disorder. These deficits include an inability to filter sensory input as measured by, e.g., prepulse inhibition (PPI) reflex. Administration of phencyclidine (PCP), a drug that can induce a schizophrenia-like psychosis in humans, disrupts PPI in experimental animals. In rodents, this PCP-induced deficit can be blocked by pretreatment with nitric oxide (NO) synthase inhibitors. This suggests that some of the behavioral effects of PCP are mediated via NO. The substrate for in vivo NO production is L-arginine, and active transport of L-arginine via the cationic amino acid transporter may serve as a regulatory mechanism in NO production. OBJECTIVES: The aim of the present study was to study the effects of L-arginine transport inhibition, using acute and repeated L-lysine treatment, on PCP-induced disruption of PPI in mice. RESULTS: Subchronic, and to some extent acute, pretreatment with L-lysine blocked a PCP-induced deficit in PPI without affecting basal PPI. CONCLUSIONS: L-lysine has been shown to block L-arginine transport in vitro, most likely via a competitive blockade and down regulation of cationic amino acid transporters. However, the importance of L-arginine transport as a regulatory mechanism in NO production in vivo is still not clear. The present results lend further support to the notion that some of the effects of PCP in the central nervous system are mediated via NO and that L-arginine transport may play a role in the regulation of NO production in the brain.


Subject(s)
Amino Acids/pharmacology , Hallucinogens/antagonists & inhibitors , Lysine/pharmacology , Phencyclidine/antagonists & inhibitors , Reflex, Startle/drug effects , Amino Acid Transport Systems, Basic/metabolism , Amino Acids/administration & dosage , Animals , Arginine/metabolism , Behavior, Animal/drug effects , Biological Transport/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Hallucinogens/pharmacology , Lysine/administration & dosage , Male , Mice , Nitric Oxide/biosynthesis , Nitric Oxide/metabolism , Phencyclidine/pharmacology , Reflex/drug effects , Schizophrenia
20.
J Psychopharmacol ; 31(12): 1588-1604, 2017 12.
Article in English | MEDLINE | ID: mdl-28946779

ABSTRACT

Enhancement of cholinergic function via muscarinic acetylcholine receptor M1 agonism improves cognition in some schizophrenia patients. Most atypical antipsychotic drugs, including clozapine and its active metabolite, N-desmethylclozapine, and lurasidone, enhance the release of acetylcholine in key brain regions involved in cognition (e.g. hippocampus). We determined the effect of muscarinic acetylcholine receptor M1 stimulation on novel object recognition and its contribution to the ability of atypical antipsychotic drugs to reverse the novel object recognition deficit in rats withdrawn from subchronic phencyclidine, a rodent model of cognitive impairment in schizophrenia. In control rats, the non-specific muscarinic acetylcholine receptor antagonist, scopolamine, and the M1 selective antagonist, VU0255035, induced a novel object recognition deficit, which was reversed by the M1 agonist, AC260584. Scopolamine fully blocked the effect of clozapine and N-desmethylclozapine, but not lurasidone, to restore novel object recognition in subchronic phencyclidine-treated rats. VU0255035 also blocked these effects of clozapine and N-desmethylclozapine, but not lurasidone; however, the blockade was not as complete as that achieved with scopolamine. Furthermore, subchronic phencyclidine increased hippocampal M1 mRNA expression. These data suggest that M1 agonism is required for clozapine and N-desmethylclozapine to ameliorate the phencyclidine-induced deficit in novel object recognition, additional evidence that M1 agonism is a potential target for treating cognitive impairment in schizophrenia.


Subject(s)
Antipsychotic Agents/pharmacology , Cognitive Dysfunction/drug therapy , Phencyclidine/antagonists & inhibitors , Phencyclidine/pharmacology , Receptor, Muscarinic M1/metabolism , Recognition, Psychology/drug effects , Signal Transduction , Animals , Benzoxazines/agonists , Clozapine/analogs & derivatives , Clozapine/pharmacology , Cognitive Dysfunction/chemically induced , Drug Interactions , Female , Lurasidone Hydrochloride/pharmacology , Rats , Receptor, Muscarinic M1/agonists , Receptor, Muscarinic M1/antagonists & inhibitors , Scopolamine/antagonists & inhibitors , Scopolamine/pharmacology , Signal Transduction/drug effects , Sulfonamides/antagonists & inhibitors , Sulfonamides/pharmacology , Thiadiazoles/antagonists & inhibitors , Thiadiazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL