Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 161
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
Psychopharmacology (Berl) ; 232(23): 4285-92, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26319159

ABSTRACT

RATIONALE: Schizophrenic-spectrum patients commonly display deficits in preattentive information processing as evidenced, for example, by disrupted prepulse inhibition (PPI), a measure of sensorimotor gating. Similar disruptions in PPI can be induced in rodents and primates by the psychotomimetic drug phencyclidine (PCP), a noncompetitive inhibitor of the NMDA receptor. Mounting evidence suggests that the hunger hormone ghrelin and its constitutively active receptor influences neuronal circuits involved in the regulation of mood and cognition. OBJECTIVES: In the present series of experiments, we investigated the effects of ghrelin and the growth hormone secretagogue receptor (GHS-R1A) neutral antagonist, JMV 2959, on acoustic startle responses (ASR), PPI, and PCP-induced alterations in PPI. RESULTS: Intraperitoneal (i.p.) administration of ghrelin (0.033, 0.1, and 0.33 mg/kg) did not alter the ASR or PPI in rats. Conversely, i.p. injection of JMV 2959 (1, 3, and 6 mg/kg), dose dependently decreased the ASR and increased PPI. Pretreatment with JMV 2959 at a dose with no effect on ASR or PPI per se, completely blocked PCP-induced (2 mg/kg) deficits in PPI while pretreatment with the highest dose of ghrelin did not potentiate or alter PPI responses of a sub-threshold dose of PCP (0.75 mg/kg). CONCLUSION: These findings indicate that the GHS-R1A is involved in specific behavioral effects of PCP and may have relevance for patients with schizophrenia.


Subject(s)
Glycine/analogs & derivatives , Phencyclidine/pharmacology , Prepulse Inhibition/drug effects , Receptors, Ghrelin/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Triazoles/pharmacology , Animals , Dose-Response Relationship, Drug , Glycine/pharmacology , Male , Phencyclidine/antagonists & inhibitors , Prepulse Inhibition/physiology , Rats , Rats, Sprague-Dawley , Receptors, Ghrelin/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Reflex, Startle/drug effects , Reflex, Startle/physiology , Schizophrenia/drug therapy , Sensory Gating/drug effects , Sensory Gating/physiology , Signal Transduction/drug effects , Signal Transduction/physiology
8.
Neuropsychopharmacology ; 40(3): 601-13, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25120077

ABSTRACT

Blonanserin differs from currently used serotonin 5-HT2A/dopamine-D2 receptor antagonists in that it exhibits higher affinity for dopamine-D2/3 receptors than for serotonin 5-HT2A receptors. We investigated the involvement of dopamine-D3 receptors in the effects of blonanserin on cognitive impairment in an animal model of schizophrenia. We also sought to elucidate the molecular mechanism underlying this involvement. Blonanserin, as well as olanzapine, significantly ameliorated phencyclidine (PCP)-induced impairment of visual-recognition memory, as demonstrated by the novel-object recognition test (NORT) and increased extracellular dopamine levels in the medial prefrontal cortex (mPFC). With blonanserin, both of these effects were antagonized by DOI (a serotonin 5-HT2A receptor agonist) and 7-OH-DPAT (a dopamine-D3 receptor agonist), whereas the effects of olanzapine were antagonized by DOI but not by 7-OH-DPAT. The ameliorating effect was also antagonized by SCH23390 (a dopamine-D1 receptor antagonist) and H-89 (a protein kinase A (PKA) inhibitor). Blonanserin significantly remediated the decrease in phosphorylation levels of PKA at Thr(197) and of NR1 (an essential subunit of N-methyl-D-aspartate (NMDA) receptors) at Ser(897) by PKA in the mPFC after a NORT training session in the PCP-administered mice. There were no differences in the levels of NR1 phosphorylated at Ser(896) by PKC in any group. These results suggest that the ameliorating effect of blonanserin on PCP-induced cognitive impairment is associated with indirect functional stimulation of the dopamine-D1-PKA-NMDA receptor pathway following augmentation of dopaminergic neurotransmission due to inhibition of both dopamine-D3 and serotonin 5-HT2A receptors in the mPFC.


Subject(s)
Phencyclidine/antagonists & inhibitors , Piperazines/pharmacology , Piperidines/pharmacology , Prefrontal Cortex/drug effects , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D3/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Recognition, Psychology/drug effects , Serotonin Receptor Agonists/pharmacology , Amphetamines/pharmacology , Animals , Benzazepines/pharmacology , Benzodiazepines/antagonists & inhibitors , Benzodiazepines/pharmacology , Dopamine/metabolism , Dopamine Agonists/pharmacology , Isoquinolines/pharmacology , Male , Mice , Olanzapine , Phencyclidine/pharmacology , Phosphorylation/drug effects , Prefrontal Cortex/metabolism , Protein Kinase Inhibitors/pharmacology , Sulfonamides/pharmacology , Tetrahydronaphthalenes/pharmacology
9.
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
10.
Psychopharmacology (Berl) ; 231(16): 3151-67, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24577516

ABSTRACT

Here, we present the pharmacological characterisation of Lu AF64280, a novel, selective, brain penetrant phosphodiesterase (PDE) 2A inhibitor, in in vitro/in vivo assays indicative of PDE2A inhibition, and in vivo models/assays relevant to cognitive processing or antipsychotic-like activity. The in vitro selectivity of Lu AF64280 was determined against a panel of PDE enzymes and 3',5'-cyclic guanosine monophosphate (cGMP) levels in the hippocampus were determined using in vivo microdialysis. Lu AF64280 potently inhibited hPDE2A (Ki = 20 nM), 50-fold above moderate inhibition of both hPDE9A (Ki = 1,000 nM) and hPDE10A (Ki = 1,800 nM), and displayed a >250-fold selectivity over all other full-length human recombinant PDE family members (Ki above 5,000 nM). Lu AF64280 (20 mg/kg) significantly increased cGMP levels in the hippocampus (p < 0.01 versus vehicle-treated mice), attenuated sub-chronic phencyclidine-induced deficits in novel object exploration in rats (10 mg/kg, p < 0.001 versus vehicle-treated), blocked early postnatal phencyclidine-induced deficits in the intradimensional/extradimensional shift task in rats (1 and 10 mg/kg, p < 0.001 versus vehicle-treated) and attenuated spontaneous P20-N40 auditory gating deficits in DBA/2 mice (20 mg/kg, p < 0.05 versus vehicle-treated). In contrast, Lu AF64280 failed to attenuate phencyclidine-induced hyperactivity in mice, and was devoid of antipsychotic-like activity in the conditioned avoidance response paradigm in rats, at any dose tested. Lu AF64280 represents a novel tool compound for selective PDE2A inhibition that substantiates a critical role of this enzyme in cognitive processes under normal and pathological conditions.


Subject(s)
Antipsychotic Agents/pharmacology , Cognition Disorders/etiology , Cognition Disorders/psychology , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Heterocyclic Compounds, 4 or More Rings/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Schizophrenia/complications , Schizophrenia/drug therapy , Schizophrenic Psychology , Animals , Antipsychotic Agents/pharmacokinetics , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacokinetics , Hippocampus/metabolism , Humans , Hyperkinesis/chemically induced , Hyperkinesis/prevention & control , Male , Mice , Mice, Inbred DBA , Phencyclidine/antagonists & inhibitors , Phencyclidine/pharmacology , Phosphodiesterase Inhibitors/pharmacokinetics , Rats , Recognition, Psychology/drug effects , Recombinant Proteins , Sensory Gating/drug effects
11.
Neuropsychopharmacology ; 38(9): 1816-24, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23563893

ABSTRACT

The neuronal mechanisms underlying social withdrawal, one of the core negative symptoms of schizophrenia, are not well understood. Recent studies suggest an involvement of the endocannabinoid system in the pathophysiology of schizophrenia and, in particular, of negative symptoms. We used biochemical, pharmacological, and behavioral approaches to investigate the role played by the endocannabinoid system in social withdrawal induced by sub-chronic administration of phencyclidine (PCP). Pharmacological enhancement of endocannabinoid levels via systemic administration of URB597, an inhibitor of endocannabinoid degradation, reversed social withdrawal in PCP-treated rats via stimulation of CB1 receptors, but reduced social interaction in control animals through activation of a cannabinoid/vanilloid-sensitive receptor. In addition, the potent CB agonist CP55,940 reversed PCP-induced social withdrawal in a CB1-dependent manner, whereas pharmacological blockade of CB1 receptors by either AM251 or SR141716 reduced the time spent in social interaction in control animals. PCP-induced social withdrawal was accompanied by a decrease of anandamide (AEA) levels in the amygdala and prefrontal cortex, and these deficits were reversed by URB597. As CB1 receptors are predominantly expressed on GABAergic interneurons containing the anxiogenic peptide cholecystokinin (CCK), we also examined whether the PCP-induced social withdrawal resulted from deficient CB1-mediated modulation of CCK transmission. The selective CCK2 antagonist LY225910 blocked both PCP- and AM251-induced social withdrawal, but not URB597 effect in control rats. Taken together, these findings indicate that AEA-mediated activation of CB1 receptors is crucial for social interaction, and that PCP-induced social withdrawal results from deficient endocannabinoid transmission.


Subject(s)
Endocannabinoids/physiology , Phencyclidine/pharmacology , Receptor, Cannabinoid, CB1/physiology , Schizophrenia/physiopathology , Social Behavior , Amidohydrolases/antagonists & inhibitors , Amygdala/drug effects , Amygdala/metabolism , Animals , Arachidonic Acids/metabolism , Benzamides/pharmacology , Cannabinoid Receptor Agonists/pharmacology , Cannabinoid Receptor Antagonists/pharmacology , Carbamates/pharmacology , Cyclohexanols/pharmacology , Dose-Response Relationship, Drug , Endocannabinoids/metabolism , Male , Phencyclidine/antagonists & inhibitors , Piperidines/antagonists & inhibitors , Piperidines/pharmacology , Polyunsaturated Alkamides/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Pyrazoles/antagonists & inhibitors , Pyrazoles/pharmacology , Quinazolinones/pharmacology , Rats , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cholecystokinin B/antagonists & inhibitors , Rimonabant
12.
Behav Brain Res ; 244: 15-28, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23384714

ABSTRACT

Reasoning and problem solving deficits have been reported in schizophrenic patients. In the present study, we have tested rats in a two-lever reversal learning task in a Skinner box to model these deficits. In other studies using the Skinner box, atypical antipsychotics fully reversed phencyclidine (PCP)-induced impairments in reversal learning which is in contrast to clinical observations where antipsychotics lack the ability to fully reverse cognitive deficits in schizophrenia. Therefore, it can be argued that the outcome of these tests may lack predictive value. In the present study, after training on a spatial discrimination between two levers, rats were exposed to a reversal of the previously learned stimulus-response contingency during 5 days. We first investigated the effects of sub-chronic treatment with the non-competitive N-methyl-d-aspartate (NMDA) antagonists dizocilpine (MK-801) and PCP on reversal learning and extinction in male Sprague Dawley rats. Subsequently, we studied the effects of different PCP treatment regimes. Then, we investigated whether the atypical antipsychotics risperidone and clozapine and the 5-hydroxytryptamine6 (5-HT6) antagonist GSK-742457 could reverse the PCP-induced deficits. All drugs were administered subcutaneously (s.c.). MK-801 did not impair reversal learning, while PCP (1.0 and 2.0 mg/kg) induced a clear deficit in reversal learning. Both compounds, however, disrupted extinction at all tested doses. Risperidone and clozapine were both ineffective in significantly ameliorating the PCP-induced deficit in reversal learning which fits well with the clinical observations. The lowest dose of clozapine (1.25 mg/kg) had an intermediate effect in ameliorating the deficit in reversal learning induced by PCP (not different from control or PCP-treated rats). The lowest dose of GSK-742457 (0.63 mg/kg) fully reversed the PCP-induced deficits while the higher dose (5.0 mg/kg) had an intermediate effect.


Subject(s)
Clozapine/pharmacology , Conditioning, Operant/drug effects , Phencyclidine/pharmacology , Quinolines/pharmacology , Reversal Learning/drug effects , Risperidone/pharmacology , Sulfones/pharmacology , Animals , Antipsychotic Agents/pharmacology , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Extinction, Psychological/drug effects , Learning Disabilities/chemically induced , Male , Phencyclidine/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Serotonin Antagonists/pharmacology
13.
Neuropharmacology ; 66: 114-21, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22445601

ABSTRACT

Despite the potential therapeutic relevance of group II metabotropic glutamate (mGlu) receptors, there has been a lack of pharmacological tools for separating the roles of mGlu2 and mGlu3 receptor subtypes. LY541850 was claimed from human mGlu receptors expressed in non-neuronal cells to be a selective orthosteric mGlu2 agonist and mGlu3 antagonist. We have verified this pharmacological profile of LY541850 in hippocampal slices. Field excitatory post-synaptic potentials (fEPSPs) evoked by stimulation of the temporo-ammonic path (TAP) input to CA1 stratum lacunosum moleculare (SLM) were inhibited by LY541850 in mGlu3-/- mice (EC(50) 38 nM) and wild-type littermates (EC(50) 42 nM) to a similar extent but were not significantly affected in mGlu2-/- mice. The group II agonist, DCG-IV, inhibited the fEPSP in all three genotypes. Co-application of DCG-IV and LY541850 in mGlu3-/- and wild-type littermates resulted in an additive effect, whereas in mGlu2-/- mice, LY541850 reversed the inhibitory action of DCG-IV. These results confirm the selective mGlu2 agonist and mGlu3 antagonist actions of LY541850. A similar profile of activity was seen in medial perforant path synapse to the dentate gyrus. Systemic administration of LY541850 to wild-type mice, reduced the increase in locomotor activity following both phencyclidine and amphetamine administration. These data support the hypothesis that mGlu2 receptors mediate the antipsychotic effects of mixed group II agonists. This article is part of a Special Issue entitled 'Metabotropic Glutamate Receptors'.


Subject(s)
Amino Acids, Dicarboxylic/pharmacology , Bridged Bicyclo Compounds/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/physiology , Receptors, Metabotropic Glutamate/physiology , Amphetamine/antagonists & inhibitors , Amphetamine/pharmacology , Animals , Cyclopropanes/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Excitatory Postsynaptic Potentials/drug effects , Glycine/analogs & derivatives , Glycine/pharmacology , Hippocampus/drug effects , Hippocampus/physiology , Mice , Mice, Inbred ICR , Mice, Knockout , Motor Activity/drug effects , Motor Activity/physiology , Phencyclidine/antagonists & inhibitors , Phencyclidine/pharmacology , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/genetics , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
14.
Schizophr Bull ; 38(5): 942-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22899397

ABSTRACT

Here, we describe our collaborative efforts to use N-methyl-d-aspartate (NMDA) receptor antagonists as a translational tool to advance our understanding of the pathophysiology of schizophrenia and identify potential new targets for treatment of schizophrenia. We began these efforts in the late 1980s with a keen sense that, in both human and animal studies, we needed to move beyond the dopamine hypothesis of schizophrenia; if the dopamine hypothesis were correct, the existing dopamine antagonists should have cured the disease but they have not. We used NMDA receptor antagonists, not to produce schizophrenia, but as a tool to provide insights into effects of disturbances in glutamate synaptic function in schizophrenia. Our work has provided insights into potential mechanisms that may contribute to disrupted cortical function in schizophrenia and has helped identify potential treatment targets for the disorder. The translational nature of this study made the clinical testing of the first of these targets feasible. Advances in systems neuroscience approaches in animals and humans make new types of translational research possible; however, our concern is that the current obstacles facing translational research funding and academia-industry collaborations threaten the future progress in this field.


Subject(s)
Neurosciences , Phencyclidine/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/physiology , Schizophrenia/drug therapy , Schizophrenia/physiopathology , Translational Research, Biomedical , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Brief Psychiatric Rating Scale , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Disease Models, Animal , Dopamine/metabolism , Emotions/drug effects , Emotions/physiology , Glutamic Acid/physiology , Humans , Ketamine/pharmacology , Phencyclidine/pharmacology , Psychoses, Substance-Induced/physiopathology , Receptor, Metabotropic Glutamate 5 , Receptors, Dopamine D2/drug effects , Receptors, Dopamine D2/physiology , Receptors, Metabotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/physiology , Synapses/drug effects , Synapses/physiology
15.
Prog Neuropsychopharmacol Biol Psychiatry ; 39(1): 192-9, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22735395

ABSTRACT

Perinatal phencyclidine (PCP) administration to rats represents one of the actual animal models of schizophrenia. Numerous data suggest redox dysregulation in this disease. We have previously demonstrated decreased content of the reduced glutathione (GSH) and complex disbalance of antioxidant enzymes in the brain of rats perinatally treated with PCP. The aim of this study was to elucidate whether chronic risperidone treatment can reverse these changes. The Wistar rats were perinatally treated with either PCP (10mg/kg; PCP, two groups) or saline (0.9% NaCl, two groups). At postnatal day (PN) 35, two groups of rats one NaCl and one PCP have started to receive risperidone in drinking water for nine weeks (NaCl-RSP and PCP-RSP groups). Animals were sacrificed on PN100 and the levels of GSH, the activities of γ-glutamate cysteine ligase (GCL), glutathione peroxidase (GPx), glutathione reductase (GR) and superoxide dismutase (SOD), as well as, the concentration of lipid peroxides were determined in the different brain structures. Risperidone restored decreased GSH levels, as well as decreased γ-GCL activity in cortex and hippocampus of animals perinatally treated with PCP. Alterations in GPx and GR activities caused by perinatal PCP treatment were also reversed by risperidone in most investigated brain structures. Furthermore, chronic risperidone treatment caused the decrease in SOD activity both in control and in PCP perinatally treated groups. Increased levels of lipid peroxides noticed in hippocampus and thalamus were reversed after chronic risperidone treatment. The results of the present study demonstrate that risperidone treatment restores GSH levels and to great measure reverses antioxidant defense alterations in the brain of perinatally PCP treated rats. Further studies are necessary in order to clarify the significance of risperidone influence on oxidative stress parameters in schizophrenia.


Subject(s)
Antioxidants/metabolism , Brain/metabolism , Glutathione/metabolism , Phencyclidine/antagonists & inhibitors , Prenatal Exposure Delayed Effects/metabolism , Risperidone/pharmacology , Schizophrenia/metabolism , Animals , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Brain/drug effects , Disease Models, Animal , Female , Glutamate-Cysteine Ligase/metabolism , Lipid Peroxidation/drug effects , Male , Pregnancy , Prenatal Exposure Delayed Effects/drug therapy , Rats , Rats, Wistar , Risperidone/therapeutic use , Schizophrenia/drug therapy
16.
Behav Brain Res ; 228(1): 211-8, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22189656

ABSTRACT

The 5-hydroxytryptamine7 (5-HT7) receptor is a G-protein coupled receptor for serotonin that has been implicated in the pathophysiology of psychiatric and neurological disorders including anxiety, depression and schizophrenia. A number of studies have attempted to evaluate the potential role of the 5-HT7 receptor in schizophrenia by utilising genetic or pharmacological tools but to date these have provided conflicting results. Here we investigate the effect of a selective 5-HT7 receptor antagonist, SB-269970, in in vivo psychosis and cognition models and relate efficacy to brain exposures of the compound. SB-269970 significantly attenuated amphetamine-induced rearing and circling in rats. A similar effect was observed in an N-methyl d-aspartic acid (NMDA) receptor antagonist driven psychosis model, where SB-269970 significantly reversed phencyclidine-induced hyperlocomotion, rearing and circling; although the effect was not as robust as with the 5-HT2a receptor antagonist positive control, MDL100,907. SB-269970 also attenuated a temporal deficit in novel object recognition (NOR), indicative of an improvement in recognition memory. Pharmacokinetic analysis of plasma and brain samples taken after behavioural testing confirmed that efficacy was achieved at doses and pre-treatment times where receptor occupancy was substantial. These findings highlight the anti-psychotic and pro-cognitive potential of 5-HT7 receptor antagonists and warrant further studies to explore their therapeutic potential in schizophrenia.


Subject(s)
Cognition/drug effects , Phenols/pharmacology , Psychotic Disorders/drug therapy , Receptors, Serotonin/drug effects , Serotonin Antagonists/pharmacology , Sulfonamides/pharmacology , Amphetamine/antagonists & inhibitors , Amphetamine/pharmacology , Animals , Animals, Outbred Strains , Disease Models, Animal , Fluorobenzenes/pharmacology , Fluorobenzenes/therapeutic use , Locomotion/drug effects , Male , Phencyclidine/antagonists & inhibitors , Phencyclidine/pharmacology , Phenols/pharmacokinetics , Phenols/therapeutic use , Piperidines/pharmacology , Piperidines/therapeutic use , Rats , Rats, Sprague-Dawley , Recognition, Psychology/drug effects , Serotonin Antagonists/pharmacokinetics , Serotonin Antagonists/therapeutic use , Sulfonamides/pharmacokinetics , Sulfonamides/therapeutic use
17.
Int Immunopharmacol ; 11(12): 2181-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22001428

ABSTRACT

Illicit drug use during pregnancy is a serious social and public health problem inflicting an array of deleterious effects on both mother and offspring. We investigated the hypothesis that a murine anti-phencyclidine (PCP) monoclonal antibody (mAb6B5; K(D)=1.3 nM) can safely protect mother and fetus from PCP-induced adverse health effects in pregnant rats. Pregnant Sprague-Dawley rats (n=4-5) were intravenously administered bolus injections of PCP (1mg/kg) on multiple days during pregnancy. They were also chronically treated with anti-PCP mAb6B5 at 45 mg/kg as a PCP antagonist. This dose provided one mAb-PCP binding site for every four PCP molecules. Therapeutic and safety study endpoints included pregnancy outcome (litter size, number of live vs. dead pups), maternal hemodynamic status and locomotor activity. Maternal hemodynamic changes (i.e., blood pressure and heart rate) and locomotor activity were measured in dams from gestation days 6-21 (one day antepartum) using a radiotelemetry-tracking device with a femoral arterial pressure catheter. This mAb6B5 treatment regimen significantly (p=0.008) reduced the number of PCP-induced in utero fetal deaths (odds ratio=3.2; 95%CI 1.3 to 7.9) and significantly (p<0.05) reduced acute PCP-induced maternal locomotor effects in the second trimester. Maternal hemodynamic responses to PCP were not significantly affected by mAb6B5 treatment. In conclusion, these data suggest that anti-PCP mAb treatments administered during pregnancy can safely protect a mother and her fetus(es) from PCP-related morbidity and mortality even when the mAb dose is too low to significantly prevent other PCP-induced maternal pharmacological effects.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Fetal Death/prevention & control , Phencyclidine/antagonists & inhibitors , Animals , Blood Pressure/drug effects , Female , Femoral Artery/drug effects , Heart Rate/drug effects , Motor Activity/drug effects , Phencyclidine/adverse effects , Pregnancy , Pregnancy Outcome , Rats
18.
J Psychopharmacol ; 25(6): 792-800, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21088042

ABSTRACT

This study investigated the effects of the dopamine D(4) receptor agonist, PD168077, on recognition memory using a novel object recognition task, which detects disruption and improvement of recognition memory in rats by measuring their ability to discriminate between familiar and novel objects. When acquisition and test were 6 h apart (experiment 1), control rats failed to discriminate between familiar and novel objects at test. Rats given low doses of PD168077 (0.3; 1.0 mg/kg) also failed to discriminate between the objects, while rats given higher doses (3.0; 10.0 mg/kg) explored the novel object more than the familiar object, indicating retained memory of the familiar object. Thus, at higher doses, PD168077 improved recognition memory in rats. Experiment 2 tested whether PD168077 would attenuate deficits in novel object recognition induced by sub-chronic phencyclidine. Testing was 1 min after acquisition, such that vehicle pre-treated rats differentiated between the novel and familiar objects: however, sub-chronic phencyclidine-treated rats failed to discriminate between the two, indicating disruption of recognition memory. PD168077 (10 mg/kg) restored the ability of phencyclidine-treated rats to differentiate between the novel and familiar objects, indicating improved recognition memory. The results suggest that D(4) receptor activation can improve cognitive dysfunction in an animal model relevant to schizophrenia.


Subject(s)
Benzamides/pharmacology , Cognition Disorders/drug therapy , Piperazines/pharmacology , Receptors, Dopamine D4/agonists , Recognition, Psychology/drug effects , Schizophrenic Psychology , Animals , Cognition Disorders/chemically induced , Cognition Disorders/complications , Discrimination, Psychological/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Female , Phencyclidine/antagonists & inhibitors , Phencyclidine/pharmacology , Rats , Rats, Inbred Strains
19.
Behav Brain Res ; 214(2): 240-7, 2010 Dec 25.
Article in English | MEDLINE | ID: mdl-20561963

ABSTRACT

BACKGROUND: Asenapine is a new pharmacological agent for the acute treatment of schizophrenia and bipolar disorder. It has relatively higher affinity for serotonergic and alpha(2)-adrenergic than dopaminergic D(2) receptors. We evaluated the effects of asenapine, risperidone, and olanzapine on acute and subchronic psychotomimetic-induced disruption of cued reversal learning in rats. METHODS: After operant training, rats were treated acutely with d-amphetamine (0.75 mg/kg intraperitoneally [i.p.]) or phencyclidine (PCP; 1.5mg/kg i.p.) or subchronically with PCP (2mg/kg i.p. for 7 days). We assessed the effects of acute coadministration of asenapine, risperidone, or olanzapine on acute d-amphetamine- and PCP-induced deficits and the effects of long-term coadministration of these agents (for 28 additional days) on the deficits induced by subchronic PCP. RESULTS: Deficits in reversal learning induced by acute d-amphetamine were attenuated by risperidone (0.2mg/kg i.p.). Acute PCP-induced impairment of reversal learning was attenuated by acute asenapine (0.025 mg/kg subcutaneously [s.c.]), risperidone (0.2mg/kg i.p.), and olanzapine (1.0mg/kg i.p.). Subchronic PCP administration induced an enduring deficit that was attenuated by acute asenapine (0.075 mg/kg s.c.) and by olanzapine (1.5mg/kg i.p.). Asenapine (0.075 mg/kg s.c.), risperidone (0.2mg/kg i.p.), and olanzapine (1.0mg/kg i.p.) all showed sustained efficacy with chronic (29 days) treatment to improve subchronic PCP-induced impairments. CONCLUSION: These data suggest that asenapine may have beneficial effects in the treatment of cognitive symptoms in schizophrenia. However, this remains to be validated by further clinical evaluation.


Subject(s)
Antipsychotic Agents/pharmacology , Benzodiazepines/pharmacology , Hallucinogens/antagonists & inhibitors , Heterocyclic Compounds, 4 or More Rings/pharmacology , Reversal Learning/drug effects , Risperidone/pharmacology , Animals , Dextroamphetamine/antagonists & inhibitors , Dextroamphetamine/pharmacology , Dibenzocycloheptenes , Drug Administration Schedule , Female , Hallucinogens/administration & dosage , Hallucinogens/pharmacology , Olanzapine , Phencyclidine/administration & dosage , Phencyclidine/antagonists & inhibitors , Phencyclidine/pharmacology , Rats , Rats, Inbred Strains
20.
Psychopharmacology (Berl) ; 210(4): 591-604, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20437030

ABSTRACT

RATIONALE: H(3)-receptor inverse agonists raise a great interest as innovative therapeutics in several central disorders. Whereas their procognitive properties are well established, their antipsychotic-like properties are still debated. OBJECTIVES: We further explored the effect of maximal doses (3-10 mg/kg) of ciproxifan, BF2.649, and ABT-239, three selective H(3)-receptor inverse agonists, on deficits of prepulse inhibition (PPI) induced by apomorphine, MK-801, and phencyclidine (PCP). Their effect was also investigated on stereotypies induced by apomorphine and methamphetamine. RESULTS: Ciproxifan, BF2.649, and ABT-239 did not reverse the PPI impairment produced by apomorphine (0.5 mg/kg, subcutaneous) in rats. Ciproxifan and BF2.649 did not reverse the impairment induced in mice by MK-801 (0.3 mg/kg). Ciproxifan and BF2.649 also failed to reverse the disruption induced in mice by PCP (5-10 mg/kg). Low to moderate doses of haloperidol (0.1-0.4 mg/kg, intraperitoneal), alone or co-administered with BF2.649, did not reverse MK-801-induced PPI disruption. A high dose (1 mg/kg) of haloperidol partially reversed the MK-801-induced deficit and BF2.649 tended to increase this effect, although nonsignificantly. Whereas stereotypies induced in mice by apomorphine and methamphetamine were totally suppressed by haloperidol, the decrease induced by ciproxifan was partial against apomorphine and very low, if any, against methamphetamine. CONCLUSIONS: Their total absence of effect in several validated animal models of the disease does not support antipsychotic properties of H(3)-receptor inverse agonists. However, their positive effects previously reported in behavioral tasks addressing learning, attention, and memory maintain the interest of H(3)-receptor inverse agonists for the treatment of cognitive symptoms of schizophrenia as adjunctive medications.


Subject(s)
Antipsychotic Agents/antagonists & inhibitors , Apomorphine/antagonists & inhibitors , Dizocilpine Maleate/antagonists & inhibitors , Drug Inverse Agonism , Inhibition, Psychological , Phencyclidine/antagonists & inhibitors , Stereotyped Behavior/drug effects , Animals , Antipsychotic Agents/pharmacology , Apomorphine/pharmacology , Benzofurans/antagonists & inhibitors , Dizocilpine Maleate/pharmacokinetics , Haloperidol/pharmacology , Histamine Antagonists/pharmacology , Imidazoles/antagonists & inhibitors , Male , Methamphetamine/antagonists & inhibitors , Methamphetamine/pharmacology , Mice , Phencyclidine/pharmacology , Piperidines/antagonists & inhibitors , Pyrrolidines/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Reflex, Startle/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...