Elsevier

Neurochemistry International

Volume 51, Issues 2–4, July–September 2007, Pages 173-184
Neurochemistry International

Review
Phencyclidine animal models of schizophrenia: Approaches from abnormality of glutamatergic neurotransmission and neurodevelopment

https://doi.org/10.1016/j.neuint.2007.06.019Get rights and content

Abstract

In humans, phencyclidine (PCP), a non-competitive N-methyl-d-aspartate (NMDA) receptor antagonist, reproduces a schizophrenia-like psychosis including positive symptoms, negative symptoms and cognitive dysfunction. Thus, the glutamatergic neuronal dysfunction hypothesis is one of the main explanatory hypotheses and PCP-treated animals have been utilized as an animal model of schizophrenia. The adult rodents treated with PCP repeatedly exhibit hyperlocomotion as an index of positive symptoms, a social behavioral deficit in a social interaction test and enhanced immobility in a forced swimming test as indices of negative symptoms. They also show a sensorimotor gating deficits and cognitive dysfunctions in several learning and memory tests. Some of these behavioral changes endure after withdrawal from repeated PCP treatment. Furthermore, repeated PCP treatment induces some neurochemical and neuroanatomical changes. On the other hand, the exposure to viral or environmental insult in the second trimester of pregnancy increases the probability of subsequently developing schizophrenia as an adult. NMDA receptor has been implicated in controlling the structure and plasticity of developing brain circuitry. Based on neurodevelopment hypothesis of schizophrenia, schizophrenia model rats treated with PCP at the perinatal stage is developed. Perinatal PCP treatment impairs neuronal development and induces long-lasting schizophrenia-like behaviors in adult period. Many findings suggest that these PCP animal models would be useful for evaluating novel therapeutic candidates and for confirming pathological mechanisms of schizophrenia.

Introduction

Schizophrenia is a severe psychiatric disease that has a lifetime prevalence of 1% in most of the populations studied (Rossler et al., 2005). Schizophrenic patients show positive symptoms (e.g., hallucinations, delusions and thought disorder), negative symptoms (e.g., deficits in social interaction, emotional expression and motivation) and cognitive dysfunction (e.g., impairments of attention and working memory) (Pearlson, 2000). The glutamatergic neuronal dysfunction hypothesis is one of the main explanatory hypotheses (Carlsson et al., 1997). It has originated from the observation that intoxication of phencyclidine (PCP), which is a non-competitive N-methyl-d-aspartate (NMDA) receptor antagonist, closely mimics schizophrenia (Javitt and Zukin, 1991). In clinical trial, acute PCP administration causes transient schizophrenic psychosis in normal volunteers and exacerbates symptoms in schizophrenic patients (Luby et al., 1959, Javitt and Zukin, 1991). Interestingly, the prolonged ingestion of PCP induces long-lasting neuropsychological deficits for several weeks (Rainey and Crowder, 1975, Allen and Young, 1978, Cosgrove and Newell, 1991).

Meanwhile, several lines of evidence suggest that the NMDA receptor is involved in the pathogenesis of schizophrenia. Postmortem studies in schizophrenic patients have identified abnormalities of NMDA receptor expression (Akbarian et al., 1996, Dracheva et al., 2001) and phosphorylation (Emamian et al., 2004) in the prefrontal cortex, which is considered to be the major region contributing to the pathophysiology of negative symptom and cognitive dysfunction in schizophrenia (Weinberger, 1988). Some genetic analyses have disclosed that single nucleotide or dinucleotide-repeated polymorphisms of the NMDA receptor subunit gene increase susceptibility to schizophrenia (Ohtsuki et al., 2001, Rice et al., 2001, Itokawa et al., 2003).

Many attempts have been made to develop animal models of schizophrenia by using NMDA receptor antagonists, such as PCP, ketamine and dizocilpine. Jentsch and Roth (1999) have proposed that the effects of chronic, rather than acute, exposure to PCP may better represent some facets of schizophrenia. In animal experiments, some of the neurochemical, neuroanatomical and behavioral alternations observed after withdrawal from repeated PCP treatment are correlated with clinical observations. Therefore, withdrawal from repeated PCP treatment might provide a valuable animal model of schizophrenia. In addition, the disorder of neurodevelopment is proposed as a trigger of the onset of schizophrenia (Lewis and Levitt, 2002, Raedler et al., 1998). Perinatal NMDA antagonist treatment developed a neurodevelopment model of schizophrenia. In this review, we focused on behavioral studies in PCP animal models of schizophrenia induced by acute and chronic treatments in adulthood and perinatal treatment and summarized recent evidence from several investigators.

Section snippets

Positive symptoms in the PCP animal model

To measure locomotor activity, animal was placed in an open field, and locomotion are measured using digital counters. PCP induces hyperlocomotion in rats and mice, which could be used as a behavioral model of the positive symptoms of schizophrenia (Sturgeon et al., 1979, Nabeshima et al., 1983), since both the first and second antipsychotics reverse the acute PCP-induced hyperlocomotion (Freed et al., 1984, Kitaichi et al., 1994). Therefore, the hyperlocomotion induced by acute PCP treatment

PCP treatment in developing period (perinatal treatment)

The first 2 weeks of postnatal life in the rat corresponding to the second trimester of pregnancy in humans, in which exposure to viral or environmental insult increases the probability of subsequently developing schizophrenia as an adult (Clancy et al., 2001). On the other hands, glutamate promotes certain aspects of neuronal development, including migration, differentiation and survival (Komuro and Rakic, 1993, Guerrini et al., 1995). NMDA receptor has been implicated in controlling the

Conclusion

Based on glutamate hypothesis, many researchers have developed PCP animal models (adult rodents treated acutely, repeatedly and perinatally with PCP). Here, we have discussed the validities of these PCP animal models for schizophrenia. Since schizophrenia has various and complex features, we have to understand the validity of PCP animal models are limited to each feature of schizophrenia and should choice suitable model for evaluating novel therapeutic candidates and for confirming pathological

Acknowledgments

This work was supported, in part, by grants-in-aid for Scientific Research on Priority Areas on “elucidation of glia-neuron network mediated information processing systems” from the Ministry of Education, Culture, Sports, Science and Technology (16047214) for scientific research from the Japan Society for the Promotion of Science (14370031, 15922139, 16922036 and 17390018), from the Research on Regulatory Science of Pharmaceuticals and Medical Devices, from a Funds from Integrated Molecular

References (101)

  • K. Hashimoto et al.

    Phencyclidine-induced cognitive deficits in mice are improved by subsequent subchronic administration of clozapine, but not haloperidol

    Eur. J. Pharmacol.

    (2005)
  • J.D. Jentsch et al.

    Subchronic phencyclidine administration reduces mesoprefrontal dopamine utilization and impairs prefrontal cortical-dependent cognition in the rat

    Neuropsychopharmacology

    (1997)
  • J.D. Jentsch et al.

    The neuropsychopharmacology of phencyclidine: from NMDA receptor hypofunction to the dopamine hypothesis of schizophrenia

    Neuropsychopharmacology

    (1999)
  • J.D. Jentsch et al.

    Subchronic phencyclidine administration increases mesolimbic dopaminergic system responsivity and augments stress- and psychostimulant-induced hyperlocomotion

    Neuropsychopharmacology

    (1998)
  • V.A. Keith et al.

    Failure of haloperidol to block the effects of phencyclidine and dizocilpine on prepulse inhibition of startle

    Biol. Psychiatry

    (1991)
  • K. Kitaichi et al.

    Effects of risperidone on phencyclidine-induced behaviors: comparison with haloperidol and ritanserin

    J. Pharmacol. Sci. (Jpn. J. Pharmacol.)

    (1994)
  • K. Kitaichi et al.

    Risperidone prevents the development of supersensitivity, but not tolerance to phencyclidine in rats treated with subacute phencyclidine

    Life Sci.

    (1995)
  • Z. Li et al.

    Effect of repeated administration of phencyclidine on spatial performance in an eight-arm radial maze with delay in rats and mice

    Pharmacol. Biochem. Behav.

    (2003)
  • B.K. Lipska et al.

    To model a psychiatric disorder in animals: schizophrenia as a reality test

    Neuropsychopharmacology

    (2000)
  • Z.A. Martinez et al.

    Effects of sustained phencyclidine exposure on sensorimotor gating of startle in rats

    Neuropsychopharmacology

    (1999)
  • J.P. Marquis et al.

    Schizophrenia-like syndrome inducing agent phencyclidine failed to impair memory for temporal order in rats

    Neurobiol. Learn. Mem.

    (2003)
  • R. Murai et al.

    Hypofunctional glutamatergic neurotransmission in the prefrontal cortex is involved in the emotional deficit induced by repeated treatment with phencyclidine in mice: implications for abnormalities of glutamate release and NMDA-CaMKII signaling

    Behav. Brain Res.

    (2007)
  • T. Nabeshima et al.

    Development of tolerance and supersensitivity to phencyclidine in rats after repeated administration of phencyclidine

    Eur. J. Pharmacol.

    (1987)
  • T. Nabeshima et al.

    Effect of lesions in the striatum, nucleus accumbens and medial raphe on phencyclidine-induced stereotyped behaviors and hyperactivity in rats

    Eur. J. Pharmacol.

    (1983)
  • A. Noda et al.

    Phencyclidine impairs latent learning in mice: interaction between glutamatergic systems and sigma1 receptors

    Neuropsychopharmacology

    (2001)
  • Y. Noda et al.

    Repeated phencyclidine treatment induces negative symptom-like behavior in forced swimming test in mice: imbalance of prefrontal serotonergic and dopaminergic functions

    Neuropsychopharmacology

    (2000)
  • Y. Noda et al.

    Effects of antidepressants on phencyclidine-induced enhancement of immobility in a forced swimming test in mice

    Eur. J. Pharmacol.

    (1997)
  • T.J. Raedler et al.

    Schizophrenia as a developmental disorder of the cerebral cortex

    Curr. Opin. Neurobiol.

    (1998)
  • L.M. Reynolds et al.

    Chronic phencyclidine administration induces schizophrenia-like changes in N-acetylaspartate and N-acetylaspartylglutamate in rat brain

    Schizophr. Res.

    (2005)
  • W. Rossler et al.

    Size of burden of schizophrenia and psychotic disorders

    Eur. Neuropsychopharmacol.

    (2005)
  • T.M. Rushe et al.

    Episodic memory and learning in patients with chronic schizophrenia

    Schizophr. Res.

    (1999)
  • C.J. Shatz

    Impulse activity and the patterning of connections during CNS development

    Neuron

    (1990)
  • R. Sircar

    Postnatal phencyclidine-induced deficit in adult water maze performance is associated with N-methyl-d-aspartate receptor upregulation

    Int. J. Dev. Neurosci.

    (2003)
  • M.R. Stefani et al.

    Effects of repeated treatment with amphetamine or phencyclidine on working memory in the rat

    Behav. Brain Res.

    (2002)
  • C. Wang et al.

    Long-term behavioral and neurodegenerative effects of perinatal phencyclidine administration: implications for schizophrenia

    Neuroscience

    (2001)
  • D.R. Weinberger

    Schizophrenia and the frontal lobe

    Trends Neurosci.

    (1988)
  • J.L. Wiley et al.

    Pharmacological challenge reveals long-term effects of perinatal phencyclidine on delayed spatial alternation in rats

    Prog. Neuropsychopharmacol. Biol. Psychiatry

    (2003)
  • Z. Abdul-Monim et al.

    Sub-chronic psychotomimetic phencyclidine induces deficits in reversal learning and alterations in parvalbumin-immunoreactive expression in the rat

    J. Psychopharmacol.

    (2006)
  • Z. Abdul-Monim et al.

    The atypical antipsychotic ziprasidone, but not haloperidol, improves phencyclidine-induced cognitive deficits in a reversal learning task in the rat

    J. Psychopharmacol.

    (2003)
  • S. Akbarian et al.

    Selective alterations in gene expression for NMDA receptor subunits in prefrontal cortex of schizophrenics

    J. Neurosci.

    (1996)
  • R.M. Allen et al.

    Phencyclidine-induced psychosis

    Am. J. Psychiatry

    (1978)
  • J.D. Andersen et al.

    Spatial memory deficits induced by perinatal treatment of rats with PCP and reversal effect of D-serine

    Neuropsychopharmacology

    (2004)
  • J.F. Artaloytia et al.

    Negative signs and symptoms secondary to antipsychotics: a double-blind, randomized trial of a single dose of placebo, haloperidol, and risperidone in healthy volunteers

    Am. J. Psychiatry

    (2006)
  • V.P. Bakshi et al.

    Antagonism of phencyclidine-induced deficits in prepulse inhibition by the putative atypical antipsychotic olanzapine

    Psychopharmacology

    (1995)
  • V.P. Bakshi et al.

    Clozapine antagonizes phencyclidine-induced deficits in sensorimotor gating of the startle response

    J. Pharmacol. Exp. Ther.

    (1994)
  • J.M. Birrell et al.

    Medial frontal cortex mediates perceptual attentional set shifting in the rat

    J. Neurosci.

    (2000)
  • D. Braff et al.

    Prestimulus effects on human startle reflex in normals and schizophrenics

    Psychophysiology

    (1978)
  • S.M. Cochran et al.

    Induction of metabolic hypofunction and neurochemical deficits after chronic intermittent exposure to phencyclidine: differential modulation by antipsychotic drugs

    Neuropsychopharmcology

    (2003)
  • J. Cosgrove et al.

    Recovery of neuropsychological functions during reduction in use of phencyclidine

    J. Clin. Psychol.

    (1991)
  • R. Depoortere et al.

    Neurochemical, electrophysiological and pharmacological profiles of the selective inhibitor of the glycine transporter-1 SSR504734, a potential new type of antipsychotic

    Neuropsychopharmacology

    (2005)
  • Cited by (229)

    View all citing articles on Scopus
    View full text