Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 307
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Pharmacol Exp Ther ; 390(1): 14-28, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38272671

ABSTRACT

Abuse of novel arylcyclohexylamines (ACX) poses risks for toxicities, including adverse neurocognitive effects. In vivo effects of ring-substituted analogs of phencyclidine (PCP), eticyclidine (PCE), and ketamine are understudied. Adult male National Institutes of Health Swiss mice were used to assess locomotor effects of PCP and its 3-OH, 3-MeO, 3-Cl, and 4-MeO analogs, PCE and its 3-OH and 3-MeO analogs, and ketamine and its deschloro and 2F-deschloro analogs, in comparison with those of methamphetamine (METH), 3,4-methylenedioxymethamphetamine (MDMA), and two benzofuran analogs of MDMA. PCP-like interoceptive effects for all of these ACXs were determined using a food-reinforced drug discrimination procedure in adult male Sprague Dawley rats. A novel operant assay of rule-governed behavior incorporating aspects of attentional set-shifting was used to profile psychosis-like neurocognitive effects of PCP and 3-Cl-PCP in rats, in comparison with cocaine and morphine. PCP-like ACXs were more effective locomotor stimulants than the amphetamines, PCE-like ACXs were as effective as the amphetamines, and ketamine-like ACXs were less effective than the amphetamines. Addition of -Cl, -OH, or -OMe at the 3-position on the aromatic ring did not impact locomotor effectiveness, but addition of -OMe at the 4-position reduced locomotor effectiveness. Lethal effects were induced by drugs with -OH at the 3-position or -OMe at the 3- or 4-position. All novel ACXs substituted at least partially for PCP, and PCP and 3-Cl-PCP elicited dose-dependent psychosis-like neurocognitive deficits in the rule-governed behavior task not observed with cocaine or morphine. Novel ACXs exhibit substantial abuse liability and toxicities not necessarily observed with their parent drugs. SIGNIFICANCE STATEMENT: Novel arylcyclohexylamine analogs of PCP, PCE, and ketamine are appearing on the illicit market, and abuse of these drugs poses risks for toxicities, including adverse neurocognitive effects. These studies demonstrate that the novel ACXs exhibit PCP-like abuse liability in the drug discrimination assay, elicit varied locomotor stimulant and lethal effects in mice, and induce psychosis-like neurocognitive effects in rats.


Subject(s)
Phencyclidine , Rats, Sprague-Dawley , Animals , Male , Mice , Phencyclidine/analogs & derivatives , Phencyclidine/toxicity , Rats , Psychoses, Substance-Induced/etiology , Cyclohexylamines , Motor Activity/drug effects , Cognition/drug effects , Conditioning, Operant/drug effects , Locomotion/drug effects , Illicit Drugs/adverse effects , Illicit Drugs/toxicity , Ketamine/analogs & derivatives , Ketamine/toxicity , Substance-Related Disorders/psychology , Phencyclidine Abuse
2.
Toxicol Appl Pharmacol ; 456: 116282, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36252887

ABSTRACT

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.


Subject(s)
Nicotine , Phencyclidine , Mice , Animals , Phencyclidine/toxicity , Nicotine/toxicity , Raclopride/pharmacology , Locomotion , Motor Activity , Receptors, Dopamine
3.
Biochem Biophys Res Commun ; 534: 610-616, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33228965

ABSTRACT

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.


Subject(s)
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
4.
Int J Mol Sci ; 22(14)2021 Jul 17.
Article in English | MEDLINE | ID: mdl-34299276

ABSTRACT

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.


Subject(s)
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
5.
J Pharmacol Exp Ther ; 371(1): 1-14, 2019 10.
Article in English | MEDLINE | ID: mdl-31371483

ABSTRACT

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.


Subject(s)
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
6.
Synapse ; 73(5): e22084, 2019 05.
Article in English | MEDLINE | ID: mdl-30582667

ABSTRACT

Schizophrenia is a mental disorder characterized by positive symptoms, negative symptoms, and cognitive dysfunction. Phencyclidine (PCP)-a N-methyl-D-aspartate (NMDA) receptor antagonist-induces symptoms indistinguishable from those of schizophrenia. A reduction of the phosphoprotein synapsin II has also been implicated in schizophrenia and has a well-known role in the maintenance of the presynaptic reserve pool and vesicle mobilization. This study assessed the behavioral and biochemical outcomes of chronic NMDA receptor antagonism in rodents and its implications for the pathophysiology of schizophrenia. Sprague Dawley rats received saline or chronic PCP (5 mg/kg/day) for 14 days via surgically implanted Alzet® osmotic mini-pumps. Following the treatment period, rats were tested with a series of behavioral paradigms, including locomotor activity, social interaction, and sensorimotor gating. Following behavioral assessment, the medial prefrontal cortex (mPFC) of all rats was isolated for synapsin II protein analysis. Chronic PCP treatment yielded a hyper-locomotive state (p = 0.0256), reduced social interaction (p = 0.0005), and reduced pre-pulse inhibition (p < 0.0001) in comparison to saline-treated controls. Synapsin IIa (p < 0.0001) and IIb (p < 0.0071) levels in the mPFC of chronically treated PCP rats were reduced in comparison to the saline group. Study results confirm that rats subject to chronic PCP treatment display behavioral phenotypes similar to established preclinical animal models of schizophrenia. Reduction of synapsin II expression in this context implicates the role of this protein in the pathophysiology of schizophrenia and sheds light on the longer-term consequences of NMDA receptor antagonism facilitated by chronic PCP treatment.


Subject(s)
Excitatory Amino Acid Antagonists/toxicity , Hallucinogens/toxicity , Phencyclidine/toxicity , Schizophrenia/metabolism , Synapsins/metabolism , Animals , Disease Models, Animal , Locomotion , Male , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , Schizophrenia/etiology , Schizophrenia/physiopathology , Social Behavior , Synapsins/genetics
7.
Proc Natl Acad Sci U S A ; 113(50): E8178-E8186, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27911814

ABSTRACT

The current dopamine (DA) hypothesis of schizophrenia postulates striatal hyperdopaminergia and cortical hypodopaminergia. Although partial agonists at DA D2 receptors (D2Rs), like aripiprazole, were developed to simultaneously target both phenomena, they do not effectively improve cortical dysfunction. In this study, we investigate the potential for newly developed ß-arrestin2 (ßarr2)-biased D2R partial agonists to simultaneously target hyper- and hypodopaminergia. Using neuron-specific ßarr2-KO mice, we show that the antipsychotic-like effects of a ßarr2-biased D2R ligand are driven through both striatal antagonism and cortical agonism of D2R-ßarr2 signaling. Furthermore, ßarr2-biased D2R agonism enhances firing of cortical fast-spiking interneurons. This enhanced cortical agonism of the biased ligand can be attributed to a lack of G-protein signaling and elevated expression of ßarr2 and G protein-coupled receptor (GPCR) kinase 2 in the cortex versus the striatum. Therefore, we propose that ßarr2-biased D2R ligands that exert region-selective actions could provide a path to develop more effective antipsychotic therapies.


Subject(s)
Antipsychotic Agents/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/metabolism , beta-Arrestin 2/metabolism , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Dopamine D2 Receptor Antagonists/pharmacology , Female , G-Protein-Coupled Receptor Kinase 2/metabolism , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Interneurons/metabolism , Ligands , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Phencyclidine/toxicity , Signal Transduction/drug effects
8.
Arch Pharm (Weinheim) ; 352(4): e1800306, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30702760

ABSTRACT

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.


Subject(s)
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
9.
J Neurosci ; 37(49): 12031-12049, 2017 12 06.
Article in English | MEDLINE | ID: mdl-29118102

ABSTRACT

We used the psychotomimetic phencyclidine (PCP) to investigate the relationships among cognitive behavior, coordinated neural network function, and information processing within the hippocampus place cell system. We report in rats that PCP (5 mg/kg, i.p.) impairs a well learned, hippocampus-dependent place avoidance behavior in rats that requires cognitive control even when PCP is injected directly into dorsal hippocampus. PCP increases 60-100 Hz medium-freguency gamma oscillations in hippocampus CA1 and these increases correlate with the cognitive impairment caused by systemic PCP administration. PCP discoordinates theta-modulated medium-frequency and slow gamma oscillations in CA1 LFPs such that medium-frequency gamma oscillations become more theta-organized than slow gamma oscillations. CA1 place cell firing fields are preserved under PCP, but the drug discoordinates the subsecond temporal organization of discharge among place cells. This discoordination causes place cell ensemble representations of a familiar space to cease resembling pre-PCP representations despite preserved place fields. These findings point to the cognitive impairments caused by PCP arising from neural discoordination. PCP disrupts the timing of discharge with respect to the subsecond timescales of theta and gamma oscillations in the LFP. Because these oscillations arise from local inhibitory synaptic activity, these findings point to excitation-inhibition discoordination as the root of PCP-induced cognitive impairment.SIGNIFICANCE STATEMENT Hippocampal neural discharge is temporally coordinated on timescales of theta and gamma oscillations in the LFP and the discharge of a subset of pyramidal neurons called "place cells" is spatially organized such that discharge is restricted to locations called a cell's "place field." Because this temporal coordination and spatial discharge organization is thought to represent spatial knowledge, we used the psychotomimetic phencyclidine (PCP) to disrupt cognitive behavior and assess the importance of neural coordination and place fields for spatial cognition. PCP impaired the judicious use of spatial information and discoordinated hippocampal discharge without disrupting firing fields. These findings dissociate place fields from spatial cognitive behavior and suggest that hippocampus discharge coordination is crucial to spatial cognition.


Subject(s)
CA1 Region, Hippocampal/drug effects , Hallucinogens/administration & dosage , Nerve Net/drug effects , Phencyclidine/administration & dosage , Spatial Behavior/drug effects , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , CA1 Region, Hippocampal/physiopathology , Hallucinogens/toxicity , Injections, Intraventricular , Locomotion/drug effects , Locomotion/physiology , Male , Nerve Net/physiopathology , Phencyclidine/toxicity , Rats , Rats, Long-Evans , Spatial Behavior/physiology
10.
J Neural Transm (Vienna) ; 125(4): 705-711, 2018 04.
Article in English | MEDLINE | ID: mdl-29270730

ABSTRACT

Diminished glutamate neurotransmission via the N-methyl-D-aspartate type glutamate receptor (NMDAR) has been considered to be involved in the pathophysiology of schizophrenia based upon the observation that the antagonists and autoantibodies of NMDAR cause positive, negative and cognitive symptomatologies similar to those of schizophrenia. The possible reduced extracellular levels of D-serine by overstimulation of the calcium-permeable α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate glutamate receptor (CP-AMPAR) following the NMDAR hypofunction-induced compensatory increase in the glutamate release could aggravate the NMDAR hypofunction in the brain of the drug- or antibody-associated psychoses and schizophrenia, because D-serine is an intrinsic coagonist for the NMDAR. To obtain an insight into the therapeutic approach to such a glutamate-linked psychotic state, we have studied the effects of the systemic administration of the CP-AMPAR-selective antagonist, IEM 1460 (N,N,N-trimethyl-5- [(tricyclo[3.3.1.13,7]dec-1-ylmethyl)amino]-1-pentanaminium bromide hydrobromide), on the hyperactivity following an injection of a schizophrenomimetic NMDAR antagonist, phencyclidine, in the mouse. The subcutaneous IEM 1460 application produced a dose-dependent inhibition of the increased movement counts after the subcutaneous injection of phencyclidine. This inhibiting influence was also seen on the hyperactivity elicited by another NMDAR antagonist, dizocilpine. Moreover, the IEM 1460 administration attenuated the ability of a schizophrenomimetic dopamine agonist, methamphetamine, to increase spontaneous movements. These findings indicate that dysregulation of the CP-AMPAR could, at least in part, be implicated in the glutamate pathology of schizophrenia and/or related psychotic symptoms and be a potential target for the development of their novel treatment.


Subject(s)
Adamantane/analogs & derivatives , Behavior, Animal/drug effects , Hyperkinesis/metabolism , Receptors, AMPA/antagonists & inhibitors , Schizophrenia , Adamantane/pharmacology , Animals , Excitatory Amino Acid Antagonists/toxicity , Hyperkinesis/chemically induced , Male , Mice , Mice, Inbred C57BL , Phencyclidine/toxicity
11.
J Neurophysiol ; 118(2): 1002-1011, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28539393

ABSTRACT

Symptoms of schizophrenia have been linked to insults during neurodevelopment such as NMDA receptor (NMDAR) antagonist exposure. In animal models, this leads to schizophrenia-like behavioral symptoms as well as molecular and functional changes within hippocampal and prefrontal regions. The aim of this study was to determine how administration of the NMDAR antagonist phencyclidine (PCP) during neurodevelopment affects functional network activity within the hippocampus and medial prefrontal cortex (mPFC). We recorded field potentials in vivo after electrical brain stem stimulation and observed a suppression of evoked theta power in ventral hippocampus, while evoked gamma power in mPFC was enhanced in rats administered with PCP neonatally. In addition, increased gamma synchrony elicited by acute administration of the NMDAR antagonist MK-801 was exaggerated in neonatal PCP animals. These data suggest that NMDAR antagonist exposure during brain development alters functional networks within hippocampus and mPFC possibly contributing to the reported behavioral symptoms of this animal model of schizophrenia.NEW & NOTEWORTHY We show that insults with a NMDA receptor antagonist during neurodevelopment lead to suppressed evoked theta oscillations in ventral hippocampus in adult rats, while evoked gamma oscillations are enhanced and hypersensitive to an acute challenge with a NMDA receptor antagonist in prefrontal cortex. These observations reveal the significance of neurodevelopmental disturbances in the evolvement of schizophrenia-like symptoms and contribute to the understanding of the functional deficits underlying aberrant behavior in this disease.


Subject(s)
Disease Models, Animal , Excitatory Amino Acid Antagonists/toxicity , Hippocampus/growth & development , Phencyclidine/toxicity , Prefrontal Cortex/growth & development , Schizophrenia/physiopathology , Animals , Animals, Newborn , Cortical Synchronization/drug effects , Cortical Synchronization/physiology , Dizocilpine Maleate/administration & dosage , Electric Stimulation , Excitatory Amino Acid Antagonists/administration & dosage , Gamma Rhythm/drug effects , Gamma Rhythm/physiology , Hippocampus/drug effects , Hippocampus/physiopathology , Male , Neural Pathways/drug effects , Neural Pathways/growth & development , Neural Pathways/physiopathology , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiopathology , Random Allocation , Rats , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Theta Rhythm/drug effects , Theta Rhythm/physiology
12.
J Pharmacol Exp Ther ; 361(1): 109-114, 2017 04.
Article in English | MEDLINE | ID: mdl-28167638

ABSTRACT

Phencyclidine (PCP), a noncompetitive N-methyl d-aspartate (NMDA) receptor antagonist, provides the most complete pharmacologic model of schizophrenia in humans and animals. Acute PCP causes hyperlocomotion, disrupts prepulse inhibition (PPI), and increases social avoidance in rats. We have previously shown that repeated treatment with the dopamine (DA) D2-like receptor agonists, quinpirole or ropinirole, prevents agonist-induced PPI disruption. In the present study, we examined whether repeated ropinirole treatment similarly attenuates the effects of PCP in a more complete model of schizophrenia symptoms and examined the effect of repeated D2-like agonist treatment on locomotion, PPI, and social interaction after acute PCP challenge. The acute effect of PCP (3.0 or 6.0 mg/kg) on locomotor activity was examined to establish a minimum effective dose. Thereafter, the effect of PCP challenge (3.0 mg/kg) on locomotor activity, PPI, and social interaction was assessed in adult male rats before or 7-10 days after termination of repeated daily treatment with ropinirole (0.1 mg/kg) or saline vehicle (0.1 ml/kg) for 28 days. Repeated ropinirole treatment attenuates PCP-induced hyperlocomotion, PPI deficits, and social avoidance. These findings suggest that repeated ropinirole treatment might affect a final common pathway that is vulnerable to both PCP- and dopamine agonist-induced behavioral disruption, thereby providing an alternative approach to block the effects of PCP.


Subject(s)
Avoidance Learning/drug effects , Hyperkinesis/chemically induced , Hyperkinesis/drug therapy , Indoles/administration & dosage , Phencyclidine/toxicity , Prepulse Inhibition/drug effects , Animals , Avoidance Learning/physiology , Dopamine Agonists/administration & dosage , Hyperkinesis/psychology , Male , Prepulse Inhibition/physiology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/physiology , Schizophrenia/chemically induced , Social Behavior , Treatment Outcome
13.
Adv Exp Med Biol ; 974: 257-261, 2017.
Article in English | MEDLINE | ID: mdl-28353244

ABSTRACT

Administration of the non-competitive N-methyl-D-aspartate (NMDA) receptor antagonist phencyclidine (PCP) to rodents has been used as the gold standard preclinical model for schizophrenia. PCP treatment induces hyperlocomotion and stereotypic behaviour, which resemble the positive symptoms of schizophrenia. In addition, proteomic studies have identified changes in proteins associated with energy metabolism and neurotransmission which are typical hallmarks of the disease. This chapter presents a protocol for the generation of this model, behavioural assessment and preparation of key bio-samples to provide the raw materials for proteomic analyses.


Subject(s)
Disease Models, Animal , Hormones/analysis , Immunoassay/methods , Nerve Tissue Proteins/analysis , Phencyclidine/toxicity , Proteomics/methods , Schizophrenia/chemically induced , Acute Disease , Animals , Brain Chemistry , Inflammation , Locomotion , Male , Rats , Rats, Sprague-Dawley , Schizophrenia/metabolism , Serum
14.
Behav Pharmacol ; 27(5): 415-21, 2016 08.
Article in English | MEDLINE | ID: mdl-26919286

ABSTRACT

The critical involvement of dopamine in cognitive processes has been well established, suggesting that therapies targeting dopamine metabolism may alleviate cognitive dysfunction. Catechol-O-methyl transferase (COMT) is a catecholamine-degrading enzyme, the substrates of which include dopamine, epinephrine, and norepinephrine. The present work illustrates the potential therapeutic efficacy of COMT inhibition in alleviating cognitive impairment. A brain-penetrant COMT inhibitor, tolcapone, was tested in normal and phencyclidine-treated rats and COMT-Val transgenic mice. In a novel object recognition procedure, tolcapone counteracted a 24-h-dependent forgetting of a familiar object as well as phencyclidine-induced recognition deficits in the rats at doses ranging from 7.5 to 30 mg/kg. In contrast, entacapone, a COMT inhibitor that does not readily cross the blood-brain barrier, failed to show efficacy at doses up to 30 mg/kg. Tolcapone at a dose of 30 mg/kg also improved novel object recognition performance in transgenic mice, which showed clear recognition deficits. Complementing earlier studies, our results indicate that central inhibition of COMT positively impacts recognition memory processes and might constitute an appealing treatment for cognitive dysfunction related to neuropsychiatric disorders.


Subject(s)
Benzophenones/pharmacology , Catechol O-Methyltransferase Inhibitors/pharmacology , Catechol O-Methyltransferase/drug effects , Cognition Disorders/drug therapy , Nitrophenols/pharmacology , Animals , Benzophenones/administration & dosage , Blood-Brain Barrier/metabolism , Brain/metabolism , Catechol O-Methyltransferase/genetics , Catechol O-Methyltransferase Inhibitors/administration & dosage , Catechols/pharmacology , Cognition Disorders/physiopathology , Dopamine/metabolism , Dose-Response Relationship, Drug , Male , Memory Disorders/drug therapy , Mice , Mice, Transgenic , Nitriles/pharmacology , Nitrophenols/administration & dosage , Phencyclidine/administration & dosage , Phencyclidine/toxicity , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Recognition, Psychology/drug effects , Tolcapone
15.
Clin Exp Pharmacol Physiol ; 43(12): 1234-1242, 2016 12.
Article in English | MEDLINE | ID: mdl-27562725

ABSTRACT

Reduced vesicular glutamate transporter 1 (VGLUT1) and 2 (VGLUT2) indicate glutamatergic hypofunction leading to cognitive impairment in schizophrenia. However, VGLUT3 involvement in cognitive dysfunction has not been reported in schizophrenia. Brahmi (Bacopa monnieri) might be a new treatment and prevention for cognitive deficits in schizophrenia by acting on cerebral VGLUT3 density. We aimed to study cognitive enhancement- and neuroprotective-effects of Brahmi on novel object recognition and cerebral VGLUT3 immunodensity in sub-chronic (2 mg/kg, Bid, ip) phencyclidine (PCP) rat model of schizophrenia. Rats were assigned to three groups for cognitive enhancement effect study: Group 1, Control; Group 2, PCP administration; Group 3, PCP+Brahmi. A neuroprotective-effect study was also carried out. Rats were again assigned to three groups: Group 1, Control; Group 2, PCP administration; Group 3, Brahmi+PCP. Discrimination ratio (DR) representing cognitive ability was obtained from a novel object recognition task. VGLUT3 immunodensity was measured in the prefrontal cortex, striatum and cornu ammonis fields 1-3 (CA1-3) using immunohistochemistry. We found reduced DR in the PCP group, which occurred alongside VGLUT3 reduction in all brain areas. PCP+Brahmi showed higher DR score with increased VGLUT3 immunodensity in the prefrontal cortex and striatum. Brahmi+PCP group showed a higher DR score with increased VGLUT3 immunodensity in the prefrontal cortex, striatum and CA1-3. We concluded that reduced cerebral VGLUT3 was involved in cognitive deficit in PCP-administrated rats. Receiving Brahmi after PCP restored cognitive deficit by increasing VGLUT3 in the prefrontal cortex and striatum. Receiving Brahmi before PCP prevented cognitive impairment by elevating VGLUT3 in prefrontal cortex, striatum and CA1-3. Therefore, Brahmi could be a new frontier of restoration and prevention of cognitive deficit in schizophrenia.


Subject(s)
Bacopa , Phencyclidine/toxicity , Plant Extracts/therapeutic use , Recognition, Psychology/physiology , Schizophrenia/metabolism , Vesicular Glutamate Transport Proteins/biosynthesis , Animals , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Male , Phencyclidine/administration & dosage , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Rats , Rats, Wistar , Recognition, Psychology/drug effects , Schizophrenia/chemically induced , Schizophrenia/drug therapy
16.
J Proteome Res ; 14(8): 3174-87, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26043028

ABSTRACT

Schizophrenia is a neuropsychiatric disorder affecting 1% of the world's population. Due to both a broad range of symptoms and disease heterogeneity, current therapeutic approaches to treat schizophrenia fail to address all symptomatic manifestations of the disease. Therefore, disease models that reproduce core pathological features of schizophrenia are needed for the elucidation of pathological disease mechanisms. Here, we employ a comprehensive global label-free liquid chromatography-mass spectrometry proteomic (LC-MS(E)) and metabonomic (LC-MS) profiling analysis combined with the targeted proteomics (selected reaction monitoring and multiplex immunoassay) of serum and brain tissues to investigate a chronic phencyclidine (PCP) rat model in which glutamatergic hypofunction is induced through noncompetitive NMDAR-receptor antagonism. Using a multiplex immunoassay, we identified alterations in the levels of several cytokines (IL-5, IL-2, and IL-1ß) and fibroblast growth factor-2. Extensive proteomic and metabonomic brain tissue profiling revealed a more prominent effect of chronic PCP treatment on both the hippocampal proteome and metabonome compared to the effect on the frontal cortex. Bioinformatic pathway analysis confirmed prominent abnormalities in NMDA-receptor-associated pathways in both brain regions, as well as alterations in other neurotransmitter systems such as kainate, AMPA, and GABAergic signaling in the hippocampus and in proteins associated with neurodegeneration. We further identified abundance changes in the level of the superoxide dismutase enzyme (SODC) in both the frontal cortex and hippocampus, which indicates alterations in oxidative stress and substantiates the apoptotic pathway alterations. The present study could lead to an increased understanding of how perturbed glutamate receptor signaling affects other relevant biological pathways in schizophrenia and, therefore, support drug discovery efforts for the improved treatment of patients suffering from this debilitating psychiatric disorder.


Subject(s)
Apoptosis/drug effects , Metabolomics/methods , Oxidative Stress/drug effects , Phencyclidine/toxicity , Proteomics/methods , Synaptic Transmission/drug effects , Animals , Chromatography, Liquid , Cytokines/blood , Cytokines/metabolism , Disease Models, Animal , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Hallucinogens/toxicity , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Male , Mass Spectrometry , Metabolome/drug effects , Proteome/metabolism , Rats, Sprague-Dawley , Schizophrenia/blood , Schizophrenia/chemically induced , Schizophrenia/metabolism , Signal Transduction/drug effects , Superoxide Dismutase/metabolism
17.
Cereb Cortex ; 24(9): 2522-32, 2014 Sep.
Article in English | MEDLINE | ID: mdl-23613110

ABSTRACT

A compromised γ-aminobutyric acid (GABA)ergic system is hypothesized to be part of the underlying pathophysiology of schizophrenia. N-methyl-D-aspartate (NMDA) receptor hypofunction during neurodevelopment is proposed to disrupt maturation of interneurons causing an impaired GABAergic transmission in adulthood. The present study examines prefrontal GABAergic transmission in adult rats administered with the NMDA receptor channel blocker, phencyclidine (PCP), for 3 days during the second postnatal week. Whole-cell patch-clamp recordings from pyramidal cells in PCP-treated rats showed a 22% reduction in the frequency of miniature inhibitory postsynaptic currents in layer II/III, but not in layer V pyramidal neurons of the prefrontal cortex. Furthermore, early postnatal PCP treatment caused insensitivity toward effects of the GABA transporter 1 (GAT-1) inhibitor, 1,2,5,6-tetrahydro-1-[2-[[(diphenyl-methylene)amino]oxy]ethyl]-3-pyridinecarboxylic acid, and also diminished currents passed by δ-subunit-containing GABAA receptors in layer II/III pyramidal neurons. The observed impairments in GABAergic function are compatible with the alteration of GABAergic markers as well as cognitive dysfunction observed in early postnatal PCP-treated rats and support the hypothesis that PCP administration during neurodevelopment affects the functionality of interneurons in later life.


Subject(s)
Excitatory Amino Acid Antagonists/toxicity , Phencyclidine/toxicity , Prefrontal Cortex/drug effects , Prefrontal Cortex/growth & development , gamma-Aminobutyric Acid/metabolism , Animals , Animals, Newborn , Electric Stimulation , GABA Antagonists/pharmacology , GABA Plasma Membrane Transport Proteins/metabolism , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Interneurons/drug effects , Interneurons/physiology , Miniature Postsynaptic Potentials/drug effects , Miniature Postsynaptic Potentials/physiology , Nipecotic Acids/pharmacology , Oximes/pharmacology , Patch-Clamp Techniques , Prefrontal Cortex/physiology , Pyramidal Cells/drug effects , Pyramidal Cells/growth & development , Pyramidal Cells/physiology , Pyridazines/pharmacology , Rats, Inbred ACI , Receptors, GABA-A/metabolism , Tissue Culture Techniques
18.
J Neurosci Res ; 92(11): 1599-607, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24953757

ABSTRACT

Although both the onset of schizophrenia and human phencyclidine (PCP) abuse typically present within the interval from adolescence to early adulthood, the majority of preclinical research employing the PCP model of schizophrenia has been conducted on neonatal or adult animals. The present study was designed to evaluate the behavioral and neurochemical sequelae of subchronic exposure to PCP in adolescence. Male 35-42-day-old Sprague Dawley rats were subcutaneously administered either saline (10 ml · kg(-1) ) or PCP hydrochloride (10 mg · kg(-1) ) once daily for a period of 14 days (n = 6/group). The animals were allowed to withdraw from treatment for 2 weeks, and their social and exploratory behaviors were subsequently assessed in adulthood by using the social interaction test. To examine the effects of adolescent PCP administration on the regulation of N-methyl-D-aspartate receptors (NMDARs), quantitative autoradiography was performed on brain sections of adult, control and PCP-withdrawn rats by using 20 nM (3) H-MK-801. Prior subchronic exposure to PCP in adolescence had no enduring effects on the reciprocal contact and noncontact social behavior of adult rats. Spontaneous rearing in response to the novel testing arena and time spent investigating its walls and floor were reduced in PCP-withdrawn animals compared with control. The long-term behavioral effects of PCP occurred in the absence of persistent deficits in spontaneous locomotion or self-grooming activity and were not mediated by altered NMDAR density. Our results document differential effects of adolescent PCP administration on the social and exploratory behaviors of adult rats, suggesting that distinct neurobiological mechanisms are involved in mediating these behaviors.


Subject(s)
Behavioral Symptoms/chemically induced , Exploratory Behavior/drug effects , Hallucinogens/toxicity , Interpersonal Relations , Phencyclidine/toxicity , Receptors, N-Methyl-D-Aspartate/metabolism , Age Factors , Animals , Autoradiography , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Dizocilpine Maleate/pharmacokinetics , Excitatory Amino Acid Antagonists/pharmacokinetics , Male , Motor Activity/drug effects , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Time Factors , Tritium/pharmacokinetics
19.
Mol Psychiatry ; 18(5): 543-56, 2013 May.
Article in English | MEDLINE | ID: mdl-22641180

ABSTRACT

Schizophrenia is a chronic, severe and highly complex mental illness. Current treatments manage the positive symptoms, yet have minimal effects on the negative and cognitive symptoms, two prominent features of the disease with critical impact on the long-term morbidity. In addition, antipsychotic treatments trigger serious side effects that precipitate treatment discontinuation. Here, we show that activation of the trace amine-associated receptor 1 (TAAR1), a modulator of monoaminergic neurotransmission, represents a novel therapeutic option. In rodents, activation of TAAR1 by two novel and pharmacologically distinct compounds, the full agonist RO5256390 and the partial agonist RO5263397, blocks psychostimulant-induced hyperactivity and produces a brain activation pattern reminiscent of the antipsychotic drug olanzapine, suggesting antipsychotic-like properties. TAAR1 agonists do not induce catalepsy or weight gain; RO5263397 even reduced haloperidol-induced catalepsy and prevented olanzapine from increasing body weight and fat accumulation. Finally, TAAR1 activation promotes vigilance in rats and shows pro-cognitive and antidepressant-like properties in rodent and primate models. These data suggest that TAAR1 agonists may provide a novel and differentiated treatment of schizophrenia as compared with current medication standards: TAAR1 agonists may improve not only the positive symptoms but also the negative symptoms and cognitive deficits, without causing adverse effects such as motor impairments or weight gain.


Subject(s)
Antipsychotic Agents/therapeutic use , Body Weight/drug effects , Depression/drug therapy , Receptors, G-Protein-Coupled/agonists , Schizophrenia/complications , Schizophrenia/drug therapy , Analysis of Variance , Animals , Antipsychotic Agents/pharmacology , Attention/drug effects , Attention/physiology , Benzodiazepines/therapeutic use , Cocaine/administration & dosage , Conditioning, Operant/drug effects , Depression/etiology , Disease Models, Animal , Dopamine Uptake Inhibitors/administration & dosage , Electroencephalography , Hallucinogens/toxicity , Haloperidol/adverse effects , Humans , Macaca fascicularis , Magnetic Resonance Imaging , Male , Mental Recall/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microinjections , Motor Activity/drug effects , Motor Activity/genetics , Mutation , Olanzapine , Oocytes , Oxazoles/pharmacokinetics , Phencyclidine/toxicity , Phenethylamines/pharmacokinetics , Protein Binding/drug effects , Protein Binding/genetics , Pyrrolidinones/administration & dosage , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Reinforcement, Psychology , Schizophrenia/etiology , Schizophrenia/genetics , Swimming/psychology , Telemetry , Tritium/pharmacokinetics , Xenopus
20.
Neurochem Res ; 39(12): 2385-93, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25270429

ABSTRACT

Use of phencyclidine (PCP) in rodents can mimic some aspects of schizophrenia. However, the underlying mechanism is still unclear. Growing evidence indicates that neuroinflammation plays a significant role in the pathophysiology of schizophrenia. In this study, we focused on inflammatory responses as target of PCP for inducing schizophrenia-like symptoms. 3-month-old C57BL/6J mice received daily injections of PCP (20 mg/kg, i.p.) or saline for one week. PCP-injected mice produced schizophrenia-like behaviours including impaired spatial short-term memory assessed by the Y-maze task and sensorimotor gating deficits in a prepulse inhibition task. Simultaneously, chronic PCP administration induced astrocyte and microglial activation in both the cortex and hippocampus. Additionally, the proinflammatory cytokine interleukin-1ß was significantly up-regulated in PCP administrated mice. Furthermore, PCP treatment decreased ratio of the phospho-Ser9 epitope of glycogen synthase kinase-3ß (GSK3ß) over total GSK3ß, which is indicative of increased GSK3ß activity. These data demonstrate that chronic PCP in mouse produces inflammatory responses and GSK3ß activation.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Inflammation/chemically induced , Phencyclidine/toxicity , Animals , Enzyme Activation , Glycogen Synthase Kinase 3 beta , Mice
SELECTION OF CITATIONS
SEARCH DETAIL