Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 304
1.
Br J Pharmacol ; 180(2): 161-173, 2023 01.
Article En | MEDLINE | ID: mdl-36106568

BACKGROUND AND PURPOSE: Improvement of cognitive deficits in schizophrenia remains an unmet need owing to the lack of new therapies and drugs. Recent studies have reported that fingolimod, an immunomodulatory drug for treating multiple sclerosis, demonstrates anti-inflammatory and neuroprotective effects in several neurological disease models. This suggests its usefulness for ameliorating cognitive dysfunction in schizophrenia. Herein, we assessed the efficacy profile and mechanism of fingolimod in a rat model of phencyclidine (PCP)-induced schizophrenia. EXPERIMENTAL APPROACH: Male Sprague-Dawley rats were treated with PCP for 14 days. The therapeutic effect of fingolimod on cognitive function was assessed using the Morris water maze and fear conditioning tests. Hippocampal neurogenesis and the expression of astrocytes and microglia were evaluated using immunostaining. Cytokine expression was quantified using multiplexed flow cytometry. Brain-derived neurotrophic factor expression and phosphorylation of extracellular signal-regulated kinase were determined using western blot analysis. KEY RESULTS: Fingolimod attenuated cognitive deficits and restored hippocampal neurogenesis in a dose-dependent manner in PCP-treated rats. Fingolimod treatment exerted anti-inflammatory effects by inhibiting microglial activation and IL-6 and IL-1ß pro-inflammatory cytokine expression. The underlying mechanism involves the upregulation of brain-derived neurotrophic factor protein expression and activation of the ERK signalling pathway. CONCLUSION AND IMPLICATIONS: This is the first preclinical assessment of the effects of fingolimod on cognitive function in a model for schizophrenia. Our results suggest the immune system plays an crucial role in cognitive alterations in schizophrenia and highlight the potential of immunomodulatory strategies to improve cognitive deficits in schizophrenia.


Cognitive Dysfunction , Schizophrenia , Animals , Rats , Male , Phencyclidine/toxicity , Schizophrenia/chemically induced , Schizophrenia/drug therapy , Schizophrenia/metabolism , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Brain-Derived Neurotrophic Factor , Rats, Sprague-Dawley , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Cytokines , Disease Models, Animal
2.
Toxicol Appl Pharmacol ; 456: 116282, 2022 12 01.
Article En | MEDLINE | ID: mdl-36252887

The association between schizophrenia and nicotine addiction becomes evident during adolescence. Here, to investigate interactive events that might underlie the early establishment of this comorbidity, we used phencyclidine-evoked locomotor sensitization, a proxy model of psychotic behavior, and nicotine minipump infusions in adolescent mice. Considering the involvement of dopamine D2 receptors in both schizophrenia and addiction, we further tested their role by exposing mice to raclopride. Adolescent mice that were either exposed to nicotine (24 mg/Kg/day) or not, received single daily raclopride (0.5 mg/kg, s.c.) or saline followed by phencyclidine injections (10 mg/Kg, s.c.) during open field testing for 6 consecutive days (Acquisition phase, ACQ). Phencyclidine and nicotine challenges (Sensitization Test, ST) were carried out after a 5-day withdrawal. Ambulation escalated in response to repeated phencyclidine exposure during ACQ and was increased after phencyclidine challenge, evidencing development and expression of locomotor sensitization. Raclopride prevented phencyclidine-evoked development of sensitization. However, raclopride pre-exposure during ACQ only shortened its expression in phencyclidine-challenged mice. Nicotine failed to interfere with phencyclidine stimulatory effects during ACQ but potentiated raclopride inhibition during the first ACQ days. During ST, nicotine history shortened the expression of phencyclidine-evoked sensitization. Nicotine challenge had no impact on locomotion, which is consistent with a lack of nicotine/phencyclidine cross-sensitization. In conclusion, our results show that nicotine does not worsen, and may even ameliorate phencyclidine-sensitized psychotic-like behavior in adolescent mice. The potentiation of raclopride-mediated inhibition further suggests that nicotine transiently improves the therapeutic efficacy of medication on psychotic symptoms through mechanisms that converge on D2 receptors.


Nicotine , Phencyclidine , Mice , Animals , Phencyclidine/toxicity , Nicotine/toxicity , Raclopride/pharmacology , Locomotion , Motor Activity , Receptors, Dopamine
3.
Int J Mol Sci ; 22(14)2021 Jul 17.
Article En | MEDLINE | ID: mdl-34299276

1-cyclohexyl-x-methoxybenzene is a novel psychoactive substance (NPS), first discovered in Europe in 2012 as unknown racemic mixture of its three stereoisomers: ortho, meta and para. Each of these has structural similarities with the analgesic tramadol and the dissociative anesthetic phencyclidine. In light of these structural analogies, and based on the fact that both tramadol and phencyclidine are substances that cause toxic effects in humans, the aim of this study was to investigate the in vitro and in vivo pharmacodynamic profile of these molecules, and to compare them with those caused by tramadol and phencyclidine. In vitro studies demonstrated that tramadol, ortho, meta and para were inactive at mu, kappa and delta opioid receptors. Systemic administration of the three stereoisomers impairs sensorimotor responses, modulates spontaneous motor activity, induces modest analgesia, and alters thermoregulation and cardiorespiratory responses in the mouse in some cases, with a similar profile to that of tramadol and phencyclidine. Naloxone partially prevents only the visual sensorimotor impairments caused by three stereoisomers, without preventing other effects. The present data show that 1-cyclohexyl-x-methoxybenzene derivatives cause pharmaco-toxicological effects by activating both opioid and non-opioid mechanisms and suggest that their use could potentially lead to abuse and bodily harm.


Analgesics, Opioid/toxicity , Anisoles/toxicity , Benzene Derivatives/toxicity , Hallucinogens/toxicity , Phencyclidine/toxicity , Psychotropic Drugs/toxicity , Receptors, Opioid/metabolism , Tramadol/toxicity , Analgesics, Opioid/chemistry , Animals , Anisoles/chemistry , Benzene Derivatives/chemistry , Cells, Cultured , Cricetinae , Hallucinogens/chemistry , In Vitro Techniques , Male , Mice , Mice, Inbred ICR , Models, Animal , Phencyclidine/chemistry , Psychotropic Drugs/chemistry , Tramadol/chemistry
4.
J Chem Neuroanat ; 116: 101993, 2021 10.
Article En | MEDLINE | ID: mdl-34147620

OBJECTIVE: We studied the role of the P2X7 receptor on cognitive dysfunction in a mouse model of schizophrenia. METHODS: An adult mouse model was established by treatment with phencyclidine (PCP), an N-methyl-D-aspartate (NMDA) receptor antagonist. Young mice were divided into three groups: 1) the control (saline-injected) group; 2) experimental 5 mg/kg PCP-injected group; and 3) experimental 10 mg/kg PCP-injected group. The mice were subjected to the open-field and Morris water maze tests at 7 weeks. After intraperitoneal injection of the P2X7 receptor antagonist JNJ-47965567, the behaviour tests were performed again. Samples were taken after testing. The P2X7 receptor protein and mRNA expression levels were detected by immunohistochemistry, Western blotting and PCR. RESULTS: This study revealed that the infant sub-chronic PCP mice model showed severe spatial learning and memory impairment in the Morris water maze and schizophrenia-like symptoms (hypermotor behaviour) in the open-field test. The P2X7 receptor protein was highly expressed in the sub-chronic PCP mouse model and more highly expressed in the hippocampus than the prefrontal lobe. After the P2X7 receptor was blocked with JNJ-47965567, P2X7 receptor protein and mRNA expression in the frontal lobe were significantly increased, and the spatial memory impairment and hypermotor behaviour induced by PCP were reversed. CONCLUSION: PCP-induced cognitive impairment can be significantly improved by antagonizing the P2X7 receptor. Therefore, we believe that the P2X7 receptor plays an important role in the cognition of schizophrenic-like mice.


Drug Delivery Systems/methods , Phencyclidine/toxicity , Purinergic P2X Receptor Antagonists/administration & dosage , Receptors, Purinergic P2X7/metabolism , Schizophrenia/drug therapy , Schizophrenia/metabolism , Animals , Animals, Newborn , Hallucinogens/toxicity , Injections, Subcutaneous , Male , Mice , Mice, Inbred C57BL , Niacinamide/administration & dosage , Niacinamide/analogs & derivatives , Piperazines/administration & dosage , Rodentia , Schizophrenia/chemically induced
5.
Neuropharmacology ; 186: 108454, 2021 03 15.
Article En | MEDLINE | ID: mdl-33444639

Positive modulation of cAMP signalling by phosphodiesterase (PDE) inhibitors has recently been explored as a potential target for the reversal of cognitive and behavioural deficits implicating the corticoaccumbal circuit. Previous studies show that PDE type 1 isoform B (PDE1B) inhibition may improve memory function in rodent models; however, the contribution of PDE1B inhibition to impulsivity, attentional and motivational functions as well as its neurophysiological effects have not been investigated. To address this, we recorded single unit activity in medial prefrontal cortex (mPFC) and nucleus accumbens (NAc) in Lister Hooded rats treated with the PDE1B inhibitor Lu AF64386 and tested in the 5-choice serial reaction time task (5-CSRTT). We also asked whether PDE1B inhibition modulates neurophysiological deficits produced by subchronic phencyclidine (PCP) treatment, a rat pharmacological model of schizophrenia. Lu AF64386 significantly affected behavioural parameters consistent with a reduction in goal-directed behaviour, however without affecting accuracy. Additionally, it reduced mPFC neuronal activity. Pre-treatment with PCP did not affect behavioural parameters, however it significantly disrupted overall neuronal firing while increasing phasic responses to reward-predicting cues and disrupting mPFC-NAc cross-talk. The latter two effects were reversed by Lu AF64386. These findings suggest PDE1B inhibition may be beneficial in disorders implicating a dysfunction of the mPFC-NAc network.


Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Goals , Phencyclidine/toxicity , Phosphodiesterase Inhibitors/therapeutic use , Prefrontal Cortex/enzymology , Schizophrenia/enzymology , Animals , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Disease Models, Animal , Female , Hallucinogens/toxicity , Phosphodiesterase Inhibitors/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiopathology , Rats , Schizophrenia/chemically induced , Schizophrenia/drug therapy
6.
Arh Hig Rada Toksikol ; 72(4): 333-342, 2021 Dec 30.
Article En | MEDLINE | ID: mdl-34985843

Lysergic acid diethylamide (LSD) is a classic hallucinogen, widely abused for decades, while phencyclidine (PCP) has increased in popularity in recent years, especially among the adolescents. Very little is known about the general toxicity of these compounds, especially about their possible neurotoxic effects at the cell level. The aim of this study was to address these gaps by assessing the toxic effects of 24-hour exposure to LSD and PCP in the concentration range of 0.39-100 µmol/L in the human neuroblastoma SH-SY5Y cell line. After cell viability was established, cells treated with concentrations that reduced their viability up to 30 % were further subjected to the alkaline comet assay and biochemical assays that enable estimation of oxidative stress-related effects. Treatment with LSD at 6.25 µmol/L and with PCP at 3.13 µmol/L resulted with 88.06±2.05 and 84.17±3.19 % of viable cells, respectively, and led to a significant increase in primary DNA damage compared to negative control. LSD also caused a significant increase in malondialdehyde level, reactive oxygen species (ROS) production, and glutathione (GSH) level, PCP significantly increased ROS but lowered GSH compared to control. Treatment with LSD significantly increased the activities of all antioxidant enzymes, while PCP treatment significantly increased superoxide dismutase (SOD) and glutathione peroxidase (GPx) but decreased catalase (CAT) activity compared to control. Our findings suggest that LSD has a greater DNA damaging potential and stronger oxidative activity than PCP in SH-SY5Y cells.


Lysergic Acid Diethylamide , Neuroblastoma , Adolescent , Cell Line , Cell Line, Tumor , DNA Damage , Humans , Lysergic Acid Diethylamide/toxicity , Oxidative Stress , Phencyclidine/toxicity , Reactive Oxygen Species , Superoxide Dismutase/metabolism
7.
Psychopharmacology (Berl) ; 238(2): 517-528, 2021 Feb.
Article En | MEDLINE | ID: mdl-33169202

RATIONALE: There is a need to develop animal models of schizophrenia-like behaviors that have both construct and predictive validity. Recently, a neonatal phencyclidine (PCP) and post-weaning social isolation dual-hit model was developed; however, its face and predictive validities need to be further investigated. OBJECTIVE: The aims of this study were to extend the characterization of the behavioral changes occurring in the neonatal PCP and post-weaning social isolation dual-hit rat model and to evaluate the effects of chronic treatment with clozapine on signs related to schizophrenia. METHODS: Male Wistar rat pups were treated with PCP (10 mg/kg s.c.) on postnatal days (PND) 7, 9, and 11. Starting from weaning, neonatal PCP-treated rat pups were socially isolated, while control saline-treated rats were group housed. At adulthood, rats were assessed using behavioral tasks evaluating locomotor activity, social recognition, prepulse inhibition, and reversal learning. Clozapine (3 mg/kg i.p.) was administered daily starting from a week before behavioral tests and until the end of the study. RESULTS: Neonatal PCP-treated and post-weaning social isolated (PCP-SI) rats displayed persistent and robust locomotor hyperactivity as well as social recognition impairment. The latter could not be explained by variations in the motivation to interact with a juvenile rat. Weak-to-moderate deficits in prepulse inhibition and reversal learning were also observed. Chronic treatment with clozapine attenuated the observed locomotor hyperactivity and social recognition deficits. CONCLUSION: The PCP-SI model presents enduring and robust deficits (hyperactivity and social recognition impairment) associated with positive symptoms and cognitive/social deficits of schizophrenia, respectively. These deficits are normalized by chronic treatment with clozapine, thereby confirming the predictive validity of this animal model.


Antipsychotic Agents/pharmacology , Clozapine/pharmacology , Executive Function/drug effects , Locomotion/drug effects , Phencyclidine/toxicity , Prepulse Inhibition/drug effects , Recognition, Psychology/drug effects , Social Isolation/psychology , Animals , Animals, Newborn , Behavior, Animal/drug effects , Disease Models, Animal , Male , Rats , Rats, Wistar , Reversal Learning/drug effects , Schizophrenia , Schizophrenic Psychology
8.
Biochem Biophys Res Commun ; 534: 610-616, 2021 01 01.
Article En | MEDLINE | ID: mdl-33228965

Schizophrenia is probably ascribed to perinatal neurodevelopmental deficits, and its onset might be affected by environmental factors. Hypofrontality with glutamatergic and dopaminergic neuronal dysfunction are known factors, but a way to mitigate abnormalities remains unfound. An early enriched environment such as a wheel running in rodents may contribute to the prevention, but its clinical applicability is very limited. From our studies, low-intensity exercise training (LET) based on physiological indices, such as lactate threshold, easily translates to humans and positively affects the brains. Hence, LET during adolescence may ameliorate abnormalities in neurodevelopment and prevent the development of schizophrenia. In the current study, LET prevented sensitization to phencyclidine (PCP) treatment, impairment of cognition, and affective behavioral abnormalities in an animal model of schizophrenia induced by prenatal PCP treatment. Further, LET increased dopamine turnover and attenuated the impairment of phosphorylation of ERK1/2 after exposure to a novel object in the prenatal PCP-treated mice. These results suggest that LET during adolescence completely improves schizophrenia-like abnormal behaviors associated with improved glutamate uptake and the dopamine-induced ERK1/2 signaling pathway in the PFC.


Physical Conditioning, Animal/methods , Schizophrenia/prevention & control , 3,4-Dihydroxyphenylacetic Acid/metabolism , Age Factors , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Dopamine/metabolism , Excitatory Amino Acid Antagonists/toxicity , Female , Homovanillic Acid/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Inbred ICR , Phencyclidine/toxicity , Phosphorylation , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/physiopathology , Prenatal Exposure Delayed Effects/psychology , Receptors, N-Methyl-D-Aspartate/metabolism , Schizophrenia/chemically induced , Schizophrenia/physiopathology , Schizophrenic Psychology
9.
Eur Neuropsychopharmacol ; 41: 92-105, 2020 12.
Article En | MEDLINE | ID: mdl-33109433

The smoking incentive in patients with schizophrenia (SCZ) depends on stimulation of nicotinic acetylcholine receptors (nAChRs) in the central nervous system. To detect potential predictor genes for nicotine responses in SCZ, we explored common factor using research data in human and animal samples. In lymphoblastoid cell lines from SCZ, the mRNA expression level of α7 nAChR subunit was decreased. In SCZ-like model mice of phencyclidine (PCP; 10 mg/kg/day, subcutaneously for 14 days)-administered mice, the mRNA expression level of α7 nAChR subunit and protein expression level of α7 or α4 nAChR subunit were significantly decreased in the prefrontal cortex during PCP withdrawal. Protein, but not mRNA, expression levels of α7, α4, and ß2 nAChR subunits were significantly increased in the nucleus accumbens. Acute (-)-nicotine [(-)-NIC: 0.3 mg/kg, s.c.] treatment attenuated impairments of social behaviors and visual recognition memory. These effects of (-)-NIC were completely blocked by both methyllycaconitine, a selective α7 nAChR antagonist, and dihydro-ß-erythroidine (DHßE), a selective α4ß2 nAChR antagonist. (-)-NIC did not induce conditioned place preference, but enhanced sensitivity to methamphetamine-induced hyperactivity. These findings suggest that α7 nAChR is associated with development of disease and is implicated in the therapeutic effect of nicotine in SCZ. The smoking incentive in SCZ might be attributed to treat their own symptoms, rather than a result of (-)-NIC dependence, by stimulating α7 and/or α4ß2 nAChRs.


Dependency, Psychological , Nicotine/therapeutic use , Phencyclidine/toxicity , Schizophrenia/chemically induced , Schizophrenia/metabolism , alpha7 Nicotinic Acetylcholine Receptor/biosynthesis , Adult , Animals , Cells, Cultured , Disease Models, Animal , Female , Humans , Male , Mice , Middle Aged , Nicotine/pharmacology , Nucleus Accumbens/chemistry , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Schizophrenia/drug therapy , Social Interaction/drug effects , alpha7 Nicotinic Acetylcholine Receptor/agonists , alpha7 Nicotinic Acetylcholine Receptor/analysis
10.
Neurochem Int ; 140: 104836, 2020 11.
Article En | MEDLINE | ID: mdl-32853750

Dopaminergic dysregulation in nucleus accumbens has been implicated in the origin of schizophrenia. Accumbal cholinergic interneurons exert powerful modulatory control of local dopamine function, through nicotinic receptors located on dopamine terminals. Fast-scan cyclic voltammetry in rat brain slices in vitro was used to measure dopamine release evoked by high-frequency electrical stimulation, mimicking phasic dopamine activity. We investigated whether cholinergic regulation of stimulated dopamine release was disrupted by pretreatment with phencyclidine, a non-competitive NMDA receptor antagonist, which provides a well validated animal model of schizophrenia. Dihydro-ß-erythroidine, an antagonist at ß2-subuit containing nicotinic receptors, caused a concentration-dependent enhancement of stimulated dopamine release, indicating cholinergic inhibitory control over dopamine release. The agonist, nicotine, also caused concentration-dependent increases in release, consistent with rapid desensitisation of the receptors previously described. In slices taken from animals pretreated with phencyclidine, the augmentation of electrically-stimulated dopamine release elicited by both drugs was attenuated, particularly when each drug was applied at high concentration. In addition, the concentration-dependence of each drug effect was lost. Taken together these findings indicate that pretreatment with phencyclidine causes changes in acetylcholine systems modulating dopamine release in accumbens. Since phencyclidine treatment was terminated at least a week before the slices were taken, the effects are due to long-term changes in function resulting from the treatment, rather than from transient changes due to the presence of the drug at test. Such enduring dysregulation of cholinergic control of phasic dopamine release could account for deficits in behaviours mediated by accumbal dopamine seen in schizophrenia, and may provide a route for novel therapeutic strategies to treat the disease.


Dopamine/metabolism , Excitatory Amino Acid Antagonists/toxicity , Nucleus Accumbens/metabolism , Phencyclidine/toxicity , Receptors, Nicotinic/metabolism , Schizophrenia/metabolism , Animals , Dihydro-beta-Erythroidine/pharmacology , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/administration & dosage , Female , Nucleus Accumbens/drug effects , Phencyclidine/administration & dosage , Rats , Rats, Wistar , Schizophrenia/chemically induced
11.
Article En | MEDLINE | ID: mdl-32061743

Autoradiography (ARG) is a high-resolution imaging method for localization of radiolabeled biomarkers in ex vivo specimen. ARG using 2-deoxy-d-glucose (2-DG) method is used in to study drug actions on brain functional activity, as it provides results comparable to clinically used functional positron-emission tomography (PET). The requirement of slow analog detection methods and emerging advances in small animal PET imaging have, however, reduced the interest in ARG. In contrast to ARG, experimental animals need to be restrained or sedated/anesthetized for PET imaging, which strongly influence functional activity and thus complicate the interpretation of the results. Digital direct particle-counting ARG systems have gained attraction during the last decade to overcome the caveats of conventional ARG methods. Here we demonstrate that the well-established 2-DG imaging method can be adapted into use with contemporary digital detectors. This method readily and rapidly captures the characteristic effects of phencyclidine (5 mg/kg, i.p.), a dissociative agent targeting the NMDAR (N-methyl-d-aspartate receptor), on regional glucose utilization in the adult mouse brain. Pretreatment with antipsychotic drug clozapine (6 mg/kg, i.p.) essentially abolishes these effects of phencyclidine on brain functional activity. Digital ARG produces viable data for the regional analysis of functional activity in a fraction of time required for film development. These results support the use of digital ARG in preclinical drug research, where high throughput and response linearity are preferred and use of sedation/anesthesia has to be avoided.


Anesthesia , Autoradiography/methods , Brain/diagnostic imaging , Brain/metabolism , Clozapine/pharmacology , Phencyclidine/toxicity , Animals , Antipsychotic Agents/pharmacology , Brain/drug effects , Hallucinogens/toxicity , Male , Mice , Mice, Inbred C57BL
13.
Pharmacol Biochem Behav ; 188: 172839, 2020 01.
Article En | MEDLINE | ID: mdl-31866390

The N-methyl-d-aspartate receptor (NMDAR) antagonists including phencyclidine (PCP) and ketamine produce cognitive deficits in rodents and humans. We previously reported that (R)-ketamine produced the beneficial effects compared to (S)-ketamine in several animal models including depression. Here we compared the effects of two enantiomers of ketamine on cognitive deficits in mice after repeated administration of PCP. PCP (10 mg/kg/day for 10 days)-induced cognitive deficits were ameliorated by subsequent repeated intermittent administration of (R)-ketamine (10 mg/kg/day, twice weekly for 2-weeks), but not (S)-ketamine. Western blot analysis showed decreased levels of brain-derived neurotrophic factor (BDNF) and decreased ratio of phosphorylated-TrkB (p-TrkB) to TrkB in the prefrontal cortex (PFC) and hippocampus of PCP-treated mice. Furthermore, PCP-induced reduction of BDNF and p-TrkB/TrkB ratio in the PFC and hippocampus of PCP-treated mice was ameliorated by subsequent intermittent administration of (R)-ketamine. Interestingly, the beneficial effects of (R)-ketamine were blocked by pretreatment with TrkB inhibitor ANA-12. These findings suggest that (R)-ketamine could ameliorate PCP-induced cognitive deficits via activation of BDNF-TrkB signaling in the brain. Therefore, (R)-ketamine could be a potential therapeutic drug for cognitive impairment in patients with schizophrenia.


Brain-Derived Neurotrophic Factor/metabolism , Cognition Disorders/drug therapy , Cognition Disorders/metabolism , Ketamine/administration & dosage , Membrane Glycoproteins/metabolism , Phencyclidine/toxicity , Protein-Tyrosine Kinases/metabolism , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Brain-Derived Neurotrophic Factor/antagonists & inhibitors , Cognition Disorders/chemically induced , Drug Administration Schedule , Excitatory Amino Acid Antagonists/administration & dosage , Male , Membrane Glycoproteins/antagonists & inhibitors , Mice , Mice, Inbred ICR , Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology
14.
J Pharmacol Exp Ther ; 371(1): 1-14, 2019 10.
Article En | MEDLINE | ID: mdl-31371483

For the past 50 years, the clinical efficacy of antipsychotic medications has relied on blockade of dopamine D2 receptors. Drug development of non-D2 compounds, seeking to avoid the limiting side effects of dopamine receptor blockade, has failed to date to yield new medicines for patients. In this work, we report the discovery of SEP-363856 (SEP-856), a novel psychotropic agent with a unique mechanism of action. SEP-856 was discovered in a medicinal chemistry effort utilizing a high throughput, high content, mouse-behavior phenotyping platform, in combination with in vitro screening, aimed at developing non-D2 (anti-target) compounds that could nevertheless retain efficacy across multiple animal models sensitive to D2-based pharmacological mechanisms. SEP-856 demonstrated broad efficacy in putative rodent models relating to aspects of schizophrenia, including phencyclidine (PCP)-induced hyperactivity, prepulse inhibition, and PCP-induced deficits in social interaction. In addition to its favorable pharmacokinetic properties, lack of D2 receptor occupancy, and the absence of catalepsy, SEP-856's broad profile was further highlighted by its robust suppression of rapid eye movement sleep in rats. Although the mechanism of action has not been fully elucidated, in vitro and in vivo pharmacology data as well as slice and in vivo electrophysiology recordings suggest that agonism at both trace amine-associated receptor 1 and 5-HT1A receptors is integral to its efficacy. Based on the preclinical data and its unique mechanism of action, SEP-856 is a promising new agent for the treatment of schizophrenia and represents a new pharmacological class expected to lack the side effects stemming from blockade of D2 signaling. SIGNIFICANCE STATEMENT: Since the discovery of chlorpromazine in the 1950s, the clinical efficacy of antipsychotic medications has relied on blockade of dopamine D2 receptors, which is associated with substantial side effects and little to no efficacy in treating the negative and cognitive symptoms of schizophrenia. In this study, we describe the discovery and pharmacology of SEP-363856, a novel psychotropic agent that does not exert its antipsychotic-like effects through direct interaction with D2 receptors. Although the mechanism of action has not been fully elucidated, our data suggest that agonism at both trace amine-associated receptor 1 and 5-HT1A receptors is integral to its efficacy. Based on its unique profile in preclinical species, SEP-363856 represents a promising candidate for the treatment of schizophrenia and potentially other neuropsychiatric disorders.


Psychotropic Drugs/pharmacology , Pyrans/pharmacology , Schizophrenia/drug therapy , Animals , Cortical Excitability/drug effects , Hallucinogens/toxicity , Macaca mulatta , Male , Mice , Mice, Inbred C57BL , Phencyclidine/toxicity , Psychotropic Drugs/therapeutic use , Pyrans/chemistry , Pyrans/therapeutic use , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, G-Protein-Coupled/agonists , Schizophrenia/etiology , Serotonin 5-HT1 Receptor Agonists/pharmacology , Serotonin 5-HT1 Receptor Agonists/therapeutic use , Sleep, REM/drug effects
15.
Neurochem Int ; 128: 127-134, 2019 09.
Article En | MEDLINE | ID: mdl-30998952

Blonanserin differs from other antipsychotic drugs, such as risperidone and olanzapine, and exhibits a higher affinity for dopamine-D2/3 receptors than for serotonin 5-HT2A receptors. We investigated the involvement of dopamine-D3 receptors in the effect of blonanserin on the social deficit observed in an animal model of schizophrenia and sought to elucidate the molecular mechanism underlying its action. Mice received phencyclidine (PCP: 10 mg/kg/day, s.c.), a non-competitive N-methyl-d-aspartate (NMDA) receptor antagonist, once a day for 14 consecutive days. We then evaluated the sociability, using a social interaction test, and the expression of GluN1 subunit, an essential subunit of the NMDA receptors, in these mice. Blonanserin significantly ameliorated the PCP-induced social deficit, whereas olanzapine and haloperidol did not. This effect of blonanserin was antagonized by 7-OH-DPAT, a dopamine-D3 receptor agonist, and SCH23390, a dopamine-D1 receptor antagonist. However, the ameliorating effect of blonanserin was not inhibited by DOI, a serotonin 5-HT2A receptor agonist. The PCP-induced social deficit was also ameliorated by U99194, a dopamine-D3 receptor antagonist and SKF38393, a dopamine-D1 receptor agonist, being effects antagonized by 7-OH-DPAT or SCH23390. Blonanserin significantly inhibited the decrease in the phosphorylation levels of GluN1 at Ser897 by protein kinase A (PKA) in the prefrontal cortex (PFC) in PCPadministered mice. These results suggest that activation of NMDA receptors due to Ser897-phosphorylation of GluN1 subunit, which is a step linked to dopamine-D1 receptor-PKA signaling through dopamine-D3 receptor antagonism in the PFC, is required for the ameliorating effect of blonanserin on the PCP-induced social deficit. These findings also provide in vivo evidence that blonanserin antagonism of the dopamine-D3 receptors may be useful as a novel treatment strategy and that the dopamine-D3 receptors can be a novel therapeutic target molecule for the social deficit observed in schizophrenia.


Dopamine Antagonists/therapeutic use , Interpersonal Relations , Phencyclidine/toxicity , Piperazines/therapeutic use , Piperidines/therapeutic use , Receptors, Dopamine D3/antagonists & inhibitors , Schizophrenia/drug therapy , Animals , Disease Models, Animal , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Hallucinogens/toxicity , Male , Mice , Mice, Inbred ICR , Piperazines/pharmacology , Piperidines/pharmacology , Schizophrenia/chemically induced
16.
Eur Neuropsychopharmacol ; 29(5): 616-628, 2019 05.
Article En | MEDLINE | ID: mdl-30910381

Schizophrenia is associated with cognitive impairments related to hypofunction in glutamatergic N-methyl-D-aspartate receptor (NMDAR) transmission. Phencyclidine (PCP), a non-competitive NMDAR antagonist, models schizophrenia-like behavioral symptoms including cognitive deficits in rodents. This study examined the effects of PCP on emotional memory function examined in the passive avoidance (PA) task in mice and the ability of typical and atypical antipsychotic drugs (APDs) to rectify the PCP-mediated impairment. Pre-training administration of PCP (0.5, 1, 2 or 3 mg/kg) dose-dependently interfered with memory consolidation in the PA task. In contrast, PCP was ineffective when administered after training, and immediately before the retention test indicating that NMDAR blockade interferes with memory encoding mechanisms. The typical APD haloperidol and the dopamine D2/3 receptor antagonist raclopride failed to block the PCP-induced PA impairment suggesting a negligible role of D2 receptors in the PCP impairment. In contrast, the memory impairment was blocked by the atypical APDs clozapine and olanzapine in a dose-dependent manner while risperidone was effective only at the highest dose tested (1 mg/kg). The PCP-induced impairment involves 5-HT1A receptor mechanisms since the antagonist NAD-299 blocked the memory impairment caused by PCP and the ability of clozapine to attenuate the impairment by PCP. These results indicate that atypical but not typical APDs can ameliorate NMDAR-mediated memory impairments and support the view that atypical APDs such as clozapine can modulate glutamatergic memory dysfunctions through 5-HT1A receptor mechanisms. These findings suggest that atypical APDs may improve cognitive impairments related to glutamatergic dysfunction relevant for emotional memories in schizophrenia.


Antipsychotic Agents/therapeutic use , Clozapine/therapeutic use , Emotional Regulation/drug effects , Haloperidol/therapeutic use , Memory Disorders/drug therapy , Phencyclidine/toxicity , Animals , Antipsychotic Agents/pharmacology , Clozapine/pharmacology , Dose-Response Relationship, Drug , Emotional Regulation/physiology , Excitatory Amino Acid Antagonists/toxicity , Haloperidol/pharmacology , Male , Memory Disorders/chemically induced , Memory Disorders/psychology , Mice , Mice, Inbred C57BL , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Serotonin 5-HT1 Receptor Antagonists/therapeutic use
17.
Psychopharmacology (Berl) ; 236(7): 2223-2232, 2019 Jul.
Article En | MEDLINE | ID: mdl-30911792

RATIONALE: Phencyclidine (PCP) could induce schizophrenia (Sz) like behavior in both humans and animals, therefore, has been widely utilized to establish Sz animal models. It induced cognitive deficits, the core symptom of Sz, mainly through influencing frontal dopaminergic function. Nonhuman primate (NHP) studies demonstrated impaired object retrieval detour (ORD) and spatial delayed response (SDR) task performance by acute or chronic PCP treatment. However, NHP investigations, continually monitoring SDR performance before, during and after PCP treatment, are lacking. OBJECTIVES: Present study investigated the long-term influence of chronic PCP treatment on SDR performance and the possible increase of SDR deficit severity and duration by the incremental dosing procedure in rhesus monkeys. METHODS: SDR task was performed repeatedly up to eight weeks after constant dosing procedure (i.m., 0.3 mg/kg, day 12-25), during which drug effects on locomotor activity and blood cortisol concentration were assessed. Incremental dosing procedure (starting dose 0.3 mg/kg, day 6-19) began five months later. RESULTS: Constant dosing procedure induced differential level of hyperactivity across testing days, without significant influence on blood cortisol concentration. It reduced SDR performance, until occurrence of the first and worst impairment on day 15 and 23 respectively. The impaired performance recovered to pretreatment level over one week after drug cessation. In contrast, incremental dosing procedure impaired SDR performance on the first treatment day, which recovered within treatment period. CONCLUSION: Results suggested increase of SDR deficit severity by repeated PCP administrations, whereas the incremental dosing procedure did not increase SDR deficit severity and duration.


Locomotion/drug effects , Memory, Short-Term/drug effects , Phencyclidine/administration & dosage , Phencyclidine/toxicity , Spatial Memory/drug effects , Animals , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/toxicity , Locomotion/physiology , Macaca mulatta , Male , Memory, Short-Term/physiology , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , Reaction Time/drug effects , Reaction Time/physiology , Schizophrenia/chemically induced , Spatial Memory/physiology
18.
Arch Pharm (Weinheim) ; 352(4): e1800306, 2019 Apr.
Article En | MEDLINE | ID: mdl-30702760

A series of benzamide derivatives possessing potent dopamine D2 , serotonin 5-HT1A , and 5-HT2A receptor properties were synthesized and evaluated as potential antipsychotics. Among them, 5-(4-(4-(benzo[d]isothiazol-3-yl)piperazin-1-yl)butoxy)-N-cyclopropyl-2-fluorobenzamide (4k) held the best pharmacological profile. It not only exhibited potent and balanced activities for the D2 , 5-HT1A , and 5-HT2A receptors, but was also endowed with low to moderate activities for the 5-HT2C , H1 , and M3 receptors, suggesting a low propensity for inducing weight gain or diabetes. In animal models, compound 4k reduced phencyclidine-induced hyperactivity with a high threshold for catalepsy or muscle relaxation induction. On the basis of its robust in vitro potency and in vivo efficacy in preclinical models of schizophrenia, 4k was selected as a candidate for further development.


Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Benzamides/pharmacology , Schizophrenia/drug therapy , Animals , Antipsychotic Agents/chemical synthesis , Antipsychotic Agents/chemistry , Benzamides/chemical synthesis , Benzamides/chemistry , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred ICR , Motor Activity/drug effects , Phencyclidine/toxicity , Receptor, Serotonin, 5-HT1A/drug effects , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT2A/drug effects , Receptor, Serotonin, 5-HT2A/metabolism , Receptors, Dopamine D2/drug effects , Receptors, Dopamine D2/metabolism , Schizophrenia/physiopathology , Structure-Activity Relationship
19.
Neurochem Int ; 125: 1-6, 2019 05.
Article En | MEDLINE | ID: mdl-30731185

Phencyclidine (PCP) is a dissociative anesthetic that induces psychotic symptoms and neurocognitive deficits in rodents similar to those observed in schizophrenia patients. PCP administration in healthy human subjects induces schizophrenia-like symptoms such as positive and negative symptoms, and a range of cognitive deficits. It has been reported that PCP, ketamine, and related drugs such as N-methyl-D-aspartate-type (NMDA) glutamate receptor antagonists, induce behavioral effects by blocking neurotransmission at NMDA receptors. Further, NMDA receptor antagonists reproduce specific aspects of the symptoms of schizophrenia. Neurochemical models based on the actions of PCP are well established, with increased focus on glutamatergic dysfunction as a basis for both symptoms and cognitive dysfunction in schizophrenia. On the other hand, the endogenous NMDA receptor antagonist, kynurenic acid (KYNA), which is a product of tryptophan-kynurenine pathway (KP) metabolism, is involved in schizophrenia pathogenesis. KYNA concentrations are elevated in the prefrontal cortex and cerebrospinal fluid of patients with schizophrenia. KYNA elevation affects neurotransmitter release in a similar manner to that of psychotomimetic agents such as PCP, underscoring a molecular basis of its involvement in schizophrenia pathophysiology. This review will highlight the relationship between PCP and KP metabolites based on evidence that both exogenous and endogenous NMDA receptor antagonists are involved in the pathogenesis of schizophrenia, and discuss our current understanding of the mechanisms underlying dysfunctional glutamatergic signaling as potential therapeutic targets for schizophrenia.


Kynurenine/metabolism , Phencyclidine/toxicity , Schizophrenia/chemically induced , Schizophrenia/metabolism , Tryptophan/metabolism , Animals , Brain/drug effects , Brain/metabolism , Humans , Signal Transduction/drug effects , Signal Transduction/physiology
20.
Psychopharmacology (Berl) ; 236(4): 1245-1253, 2019 Apr.
Article En | MEDLINE | ID: mdl-30535904

RATIONALE: Social withdrawal is a core feature of the negative symptoms of schizophrenia. Currently available pharmacotherapies have only limited efficacy towards the negative symptoms, i.e., there is a significant unmet medical need in the treatment of these symptoms. OBJECTIVE: We wanted to confirm whether selective adrenergic α2C receptor (AR) antagonist therapy could ameliorate acute phencyclidine (PCP)-induced schizophrenia-like social interaction deficits in rats, and to compare the effects of an α2C AR antagonist to another putative therapeutic alternative, an α7 nicotinic acetylcholine receptor (nAChR) partial agonist, as well against three commonly used atypical antipsychotics. METHODS: Here, we used acute PCP administration and modified a protocol for testing social interaction deficits in male Wistar rats and then used this model to compare the effects of an α2C AR antagonist (ORM-13070 0.3 and 1.0 mg/kg s.c.) with an α7 nAChR partial agonist (EVP-6124 0.3 mg/kg s.c.) and three atypical antipsychotics (clozapine 2.5 mg/kg i.p., risperidone 0.04 and 0.08 mg/kg s.c., olanzapine 0.125 and 0.5 mg/kg s.c.) on social interaction behavior. RESULTS: Acute PCP (1.5 mg/kg s.c.) produced robust and reproducible deficits in social interaction behavior without affecting locomotor activity. The selective α2C AR antagonist significantly ameliorated PCP-induced social interaction deficits. In contrast, neither the partial α7 nAChR agonist nor any of the three atypical antipsychotics were able to reverse the behavioral deficits at the selected doses. CONCLUSION: Our findings confirm that α2C AR antagonism is a potential mechanism for the treatment of the negative symptoms of schizophrenia.


Adrenergic alpha-Antagonists/therapeutic use , Interpersonal Relations , Phencyclidine/toxicity , Receptors, Adrenergic, alpha-2/physiology , Schizophrenia/chemically induced , Schizophrenia/drug therapy , Adrenergic alpha-Antagonists/pharmacology , Animals , Antipsychotic Agents/therapeutic use , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/toxicity , Male , Rats , Rats, Wistar , Social Behavior
...