Skip to main content
Log in

PCP-based mice models of schizophrenia: differential behavioral, neurochemical and cellular effects of acute and subchronic treatments

  • Original Investigation
  • Published:
Psychopharmacology Aims and scope Submit manuscript

Abstract

Rationale

N-methyl-D-aspartate receptor (NMDA-R) hypofunction has been proposed to account for the pathophysiology of schizophrenia. Thus, NMDA-R blockade has been used to model schizophrenia in experimental animals. Acute and repeated treatments have been successfully tested; however, long-term exposure to NMDA-R antagonists more likely resembles the core symptoms of the illness.

Objectives

To explore whether schizophrenia-related behaviors are differentially induced by acute and subchronic phencyclidine (PCP) treatment in mice and to examine the neurobiological bases of these differences.

Results

Subchronic PCP induced a sensitization of acute locomotor effects. Spontaneous alternation in a T-maze and novel object recognition performance were impaired after subchronic but not acute PCP, suggesting a deficit in working memory. On the contrary, reversal learning and immobility in the tail suspension test were unaffected. Subchronic PCP significantly reduced basal dopamine but not serotonin output in medial prefrontal cortex (mPFC) and markedly decreased the expression of tyrosine hydroxylase in the ventral tegmental area. Finally, acute and subchronic PCP treatments evoked a different pattern of c-fos expression. At 1 h post-treatment, acute PCP increased c-fos expression in many cortical regions, striatum, thalamus, hippocampus, and dorsal raphe. However, the increased c-fos expression produced by subchronic PCP was restricted to the retrosplenial cortex, thalamus, hippocampus, and supramammillary nucleus. Four days after the last PCP injection, c-fos expression was still increased in the hippocampus of subchronic PCP-treated mice.

Conclusions

Acute and subchronic PCP administration differently affects neuronal activity in brain regions relevant to schizophrenia, which could account for their different behavioral effects.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

References

  • Alexander GE, Crutcher MD (1990) Functional architecture of basal ganglia circuits: neural substrates of parallel processing. Trends Neurosci 13:266–271

    Article  CAS  PubMed  Google Scholar 

  • Amann LC, Gandal MJ, Halene TB, Ehrlichman RS, White SL, McCarren HS, Siegel SJ (2010) Mouse behavioral endophenotypes for schizophrenia. Brain Res Bull 83:147–161

    Article  PubMed  Google Scholar 

  • Bondi C, Matthews M, Moghaddam B (2012) Glutamatergic animal models of schizophrenia. Curr Pharm Des 18:1593–1604

    Article  CAS  PubMed  Google Scholar 

  • Boulougouris V, Glennon JC, Robbins TW (2008) Dissociable effects of selective 5-HT2A and 5-HT2C receptor antagonists on serial spatial reversal learning in rats. Neuropsychopharmacology 33:2007–2019

    Article  CAS  PubMed  Google Scholar 

  • Brigman JL, Ihne J, Saksida LM, Bussey TJ, Holmes A (2009) Effects of subchronic phencyclidine (PCP) treatment on social behaviors, and operant discrimination and reversal learning in C57BL/6J mice. Front Behav Neurosci 3:2

    Article  PubMed Central  PubMed  Google Scholar 

  • Bubser M, Schmidt WJ (1990) 6-Hydroxydopamine lesion of the rat prefrontal cortex increases locomotor activity, impairs acquisition of delayed alternation tasks, but does not affect uninterrupted tasks in the radial maze. Behav Brain Res 37:157–68

    Article  CAS  PubMed  Google Scholar 

  • Carlsson M, Carlsson A (1990) Interactions between glutamatergic and monoaminergic systems within the basal ganglia: implications for schizophrenia and Parkinson’s disease. Trends Neurosci 13:272–276

    Article  CAS  PubMed  Google Scholar 

  • Castañé A, Artigas F, Bortolozzi A (2008) The absence of 5-HT(1A) receptors has minor effects on dopamine but not serotonin release evoked by MK-801 in mice prefrontal cortex. Psychopharmacology 200:281–290

    Article  PubMed  Google Scholar 

  • Chartoff EH, Heusner CL, Palmiter RD (2005) Dopamine is not required for the hyperlocomotor response to NMDA receptor antagonists. Neuropsychopharmacology 30:1324–1333

    CAS  PubMed  Google Scholar 

  • Clarke HF, Dalley JW, Crofts HS, Robbins TW, Roberts AC (2004) Cognitive inflexibility after prefrontal serotonin depletion. Science 304:878–880

    Article  CAS  PubMed  Google Scholar 

  • Clarke HF, Walker SC, Crofts HS, Dalley JW, Robbins TW, Roberts AC (2005) Prefrontal serotonin depletion affects reversal learning but not attentional set shifting. J Neurosci 25:532–538

    Article  CAS  PubMed  Google Scholar 

  • Coyle JT (2006) Glutamate and schizophrenia: beyond the dopamine hypothesis. Cell Mol Neurobiol 26:365–384

    Article  CAS  PubMed  Google Scholar 

  • Coyle JT, Tsai G, Goff D (2003) Converging evidence of NMDA receptor hypofunction in the pathophysiology of schizophrenia. Ann N Y Acad Sci 1003:318–332

    Article  CAS  PubMed  Google Scholar 

  • Dix S, Gilmour G, Potts S, Smith JW, Tricklebank M (2010) A within-subject cognitive battery in the rat: differential effects of NMDA receptor antagonists. Psychopharmacology 212:227–242

    Article  CAS  PubMed  Google Scholar 

  • Dragunow M, Faull R (1989) The use of c-fos as a metabolic marker in neuronal pathway tracing. J Neurosci Methods 29:261–265

    Article  CAS  PubMed  Google Scholar 

  • Egerton A, Reid L, McGregor S, Cochran SM, Morris BJ, Pratt JA (2008) Subchronic and chronic PCP treatment produces temporally distinct deficits in attentional set shifting and prepulse inhibition in rats. Psychopharmacology 198:37–49

    Article  CAS  PubMed  Google Scholar 

  • Featherstone RE, Kapur S, Fletcher PJ (2007) The amphetamine-induced sensitized state as a model of schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 31:1556–1571

    Article  CAS  PubMed  Google Scholar 

  • Fellini L, Kumar G, Gibbs S, Steckler T, Talpos J (2014) Re-evaluating the PCP challenge as a pre-clinical model of impaired cognitive flexibility in schizophrenia. Eur Neuropsychopharmacol 24:1836–1849

    Article  CAS  PubMed  Google Scholar 

  • Franklin K, Paxinos G (1997) The mouse brain in stereotaxic coordinates. Academic, San Diego, California

    Google Scholar 

  • Frohlich J, Van Horn JD (2014) Reviwing the ketamine model for schizophrenia. J Psychopharmacol 28:287–330

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Gerfen CR (2000) Molecular effects of dopamine on striatal-projection pathways. Trends Neurosci 23:S64–S70

    Article  CAS  PubMed  Google Scholar 

  • Graybiel AM (1990) Neurotransmitters and neuromodulators in the basal ganglia. Trends Neurosci 13:244–254

    Article  CAS  PubMed  Google Scholar 

  • Groenewegen HJ (2003) The basal ganglia and motor control. Neural Plast 10:107–120

    Article  PubMed Central  PubMed  Google Scholar 

  • Hashimoto K, Fujita Y, Shimizu E, Iyo M (2005) Phencyclidine-induced cognitive deficits in mice are improved by subsequent subchronic administration of clozapine, but not haloperidol. Eur J Pharmacol 519:114–117

    Article  CAS  PubMed  Google Scholar 

  • Hiramatsu M, Sasaki M, Nabeshima T, Kameyama T (1997) Effects of dynorphin A (1-13) on carbon monoxide-induced delayed amnesia in mice. Pharmacol Biochem Behav 56:73–79

    Article  CAS  PubMed  Google Scholar 

  • Hiyoshi T, Kambe D, Karasawa J, Chaki S (2014) Differential effects of NMDA receptor antagonists at lower and higher doses on basal gamma band oscillation power in rat cortical electroencephalograms. Neuropharmacology 85:384–396

    Article  CAS  PubMed  Google Scholar 

  • Idris N, Neill J, Grayson B, Bang-Andersen B, Witten LM, Brennum LT, Arnt J (2010) Sertindole improves sub-chronic PCP induced reversal learning and episodic memory deficits in rodents: involvement of 5-HT(6) and 5-HT(2A) receptor mechanisms. Psychopharmacology 208:23–36

    Article  CAS  PubMed  Google Scholar 

  • Javitt DC, Zukin SR (1991) Recent advances in the phencyclidine model of schizophrenia. Am J Psychiatry 148:1301–1308

    Article  CAS  PubMed  Google Scholar 

  • Jentsch JD, Roth RH (1999) The neuropsychopharmacology of phencyclidine: from NMDA receptor hypofunction to the dopamine hypothesis of schizophrenia. Neuropsychopharmacology 20:201–225

    Article  CAS  PubMed  Google Scholar 

  • Jentsch JD, Taylor JR (2001) Impaired inhibition of conditioned responses produced by subchronic administration of phencyclidine to rats. Neuropsychopharmacology 24:66–74

    Article  CAS  PubMed  Google Scholar 

  • Jones CA, Watson DJG, Fone KCF (2011) Animal models of schizophrenia. Br J Pharmacol 164:1162–1194

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Kaffman A, Krystal JH (2012) New frontiers in animal research of psychiatric illness. Methods Mol Biol 829:3–30

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Kantrowitz JT, Javitt DC (2010) N-methyl-D-aspartate (NMDA) receptor dysfunction or dysregulation: the final common pathway on the road to schizophrenia? Brain Res Bull 83:108–121

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Kargieman L, Santana N, Mengod G, Celada P, Artigas F (2007) Antipsychotic drugs reverse the disruption in prefrontal cortex function produced by NMDA receptor blockade with phencyclidine. Proc Natl Acad Sci U S A 104:14843–14848

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Konkle AT, Bielajew C (2004) Yracing the neuroanatomical profiles of reward pathways with markers of neuronal activation. Rev Neurosci 15:383–414

    PubMed  Google Scholar 

  • Krystal JH, D’Souza DC, Mathalon D, Perry E, Belger A, Hoffman R (2003) NMDA receptor antagonist effects, cortical glutamatergic function, and schizophrenia: toward a paradigm shift in medication development. Psychopharmacology 169:215–233

    Article  CAS  PubMed  Google Scholar 

  • Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R, Bremner JD et al (1994) Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans. Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry 51:199–214

    Article  CAS  PubMed  Google Scholar 

  • Landau SM, Lal R, O’Neil JP, Baker S, Jagust WJ (2009) Striatal dopamine and working memory. Cereb Cortex 19:445–454

    Article  PubMed Central  PubMed  Google Scholar 

  • Lapin IP, Rogawski (1995) Effects of D1 and D2 dopamine receptor antagonists and catecholamine depleting agents on the locomotor stimulation induced by dizocilpine in mice. Behav Brain Res 70:145–151

    Article  CAS  PubMed  Google Scholar 

  • López Hill X, Scorza MC (2012) Role of the anterior thalamic nucleus in the motor hyperactivity induced by systemic MK-801 administration in rats. Neuropharmacology 62:2440–2446

    Article  PubMed  Google Scholar 

  • Luby ED, Cohen BD, Rosenbaum G, Gottlieb JS, Kelley R (1959) Study of a new schizophrenomimetic drug AMA. Arch Neurol Psychiatry 81:363–369

    Article  CAS  Google Scholar 

  • Ma J, Leung LS (2000) Relation between hippocampal gamma waves and behavioral disturbances induced by phencyclidine and methamphetamine. Behav Brain Res 111:1–11

    Article  CAS  PubMed  Google Scholar 

  • Ma J, Leung LS (2007) The supramammillo-septal-hippocampal pathway mediates sensorimotor gating impairment and hyperlocomotion induced by MK-801 and ketamine in rats. Psychopharmacology 191:961–974

    Article  CAS  PubMed  Google Scholar 

  • McLean SL, Woolley ML, Thomas D, Neill JC (2009) Role of 5-HT receptor mechanisms in sub-chronic PCP-induced reversal learning deficits in the rat. Psychopharmacology 206:403–414

    Article  CAS  PubMed  Google Scholar 

  • McLean SL, Neill JC, Idris NF, Marston HM, Wong EH, Shahid M (2010) Effects of asenapine, olanzapine, and risperidone on psychotomimetic-induced reversal-learning deficits in the rat. Behav Brain Res 214:240–247

    Article  CAS  PubMed  Google Scholar 

  • McLean SL, Grayson B, Idris NF, Lesage AS, Pemberton DJ, Mackie C, Neill JC (2011) Activation of alpha 7 nicotinic receptors improves phencyclidine-induced deficits in cognitive tasks inrats: implications for therapy of cognitive dysfunction in schizophrenia. Eur Neuropsychopharmacol 21:333–343

    Article  CAS  PubMed  Google Scholar 

  • Moghaddam B, Javitt D (2012) From revolution to evolution: the glutamate hypothesis of schizophrenia and its implication for treatment. Neuropsychopharmacology 37:4–15

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Mouri A, Koseki T, Narusawa S, Niwa M, Mamiya T, Kano S, Sawa A, Nabeshima T (2012) Mouse strain differences in phencyclidine-induced behavioural changes. Int J Neuropsychopharmacol 15:767–779

    Article  CAS  PubMed  Google Scholar 

  • Murphy BL, Arnsten AF, Goldman-Rakic PS, Roth RH (1996) Increased dopamine turnover in the prefrontal cortex impairs spatial working memory performance in rats and monkeys. Proc Natl Acad Sci U S A 93:1325–1329

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Nagai T, Noda Y, Une T, Furukawa K, Furukawa H, Kan QM, Nabeshima T (2003) Effect of AD-5423 on animal models of schizophrenia: phencyclidine-induced behavioral changes in mice. Neuroreport 14:269–272

    Article  CAS  PubMed  Google Scholar 

  • Neill JC, Barnes S, Cook S, Grayson B, Idris NF, McLean SL, Snigdha S, Rajagopal L, Harte MK (2010) Animal models of cognitive dysfunction and negative symptoms of schizophrenia: focus on NMDA receptor antagonism. Pharmacol Ther 128:419–432

    Article  CAS  PubMed  Google Scholar 

  • Neill JC, Harte MK, Haddad PM, Lydall ES, Dwyer DM (2014) Acute and chronic effects of NMDA receptor antagonists in rodents, relevance to negative symptoms of schizophrenia: a translational link to humans. Eur Neuropsychopharmacol 24:822–823

    Article  CAS  PubMed  Google Scholar 

  • Panagis G, Nomikos GG, Miliaressis E, Chergui K, Kastellakis A, Svensson TH, Spyraki C (1997) Ventral pallidum self-stimulation induces stimulus dependent increase in c-fos expression in reward-related brain regions. Neuroscience 77:175–186

    Article  CAS  PubMed  Google Scholar 

  • Paoletti P, Bellone C, Zhou Q (2013) NMDA receptor subunit diversity: impact on receptor properties, synaptic plasticity and disease. Nat Rev Neurosci 14:383–400

    Article  CAS  PubMed  Google Scholar 

  • Phillipson OT, Griffiths AC (1985) The topographic order of inputs to nucleus accumbens in the rat. Neuroscience 16:275–296

    Article  CAS  PubMed  Google Scholar 

  • Pierce RC, Kalivas PW (1997) A circuitry model of the expression of behavioral sensitization to amphetamine-like psychostimulants. Brain Res Brain Res Rev 25:192–221

    Article  CAS  PubMed  Google Scholar 

  • Post RM, Rose H (1976) Increasing effects of repetitive cocaine administration in the rat. Nature 260:731–732

    Article  CAS  PubMed  Google Scholar 

  • Robbins TW (2012) Animal models of neuropsychiatry revisited: a personal tribute to Teitelbaum. Behav Brain Res 231:337–342

    Article  CAS  PubMed  Google Scholar 

  • Sager SM, Sharp FR, Currant T (1988) Expression of c-fos protein in brain: metabolic mapping at the cellular level. Science 240:1328–1331

    Article  Google Scholar 

  • Santana N, Troyano-Rodriguez E, Mengod G, Celada P, Artigas F (2011) Activation of thalamocortical networks by the N-methyl-D-aspartate receptor antagonist phencyclidine: reversal by clozapine. Biol Psychiatry 69:918–927

    Article  CAS  PubMed  Google Scholar 

  • Seillier A, Giuffrida A (2009) Evaluation of NMDA receptor models of schizophrenia: divergences in the behavioral effects of subchronic PCP and MK-801. Behav Brain Res 204:410–415

    Article  CAS  PubMed  Google Scholar 

  • Simon H (1981) Dopaminergic A10 neurons and frontal system. J Physiol 77:81–95

    CAS  Google Scholar 

  • Spielewoy C, Markou A (2003) Withdrawal from chronic phencyclidine treatment induces long-lasting depression in brain reward function. Neuropsychopharmacology 28:1106–1116

    CAS  PubMed  Google Scholar 

  • Steru L, Chermat R, Thierry B, Simon P (1985) The tail suspension test: a new method for screening antidepressants in mice. Psychopharmacology 85:367–370

    Article  CAS  PubMed  Google Scholar 

  • Tanibuchi Y, Fujita Y, Kohno M, Ishima T, Takatsu Y, Iyo M, Hashimoto K (2009) Effects of quetiapine on phencyclidine-induced cognitive deficits in mice: a possible role of alpha1-adrenoceptors. Eur Neuropsychopharmacol 19:861–867

    Article  CAS  PubMed  Google Scholar 

  • Thomson DM, McVie A, Morris BJ, Pratt JA (2011) Dissociation of acute and chronic intermittent phencyclidine-induced performance deficits in the 5-choice serial reaction time task: influence of clozapine. Psychopharmacology 213:681–695

    Article  CAS  PubMed  Google Scholar 

  • Troyano-Rodríguez E, Lladó-Pelfort L, Santana N, Teruel-Martí V, Celada P, Artigas F (2014) Phencyclidine inhibits the activity of thalamic reticular gamma-aminobutyric acidergic neurons in rat brain. Biol Psychiatry 76:937–945

    Article  PubMed  Google Scholar 

  • Väisänen J, Ihalainen J, Tanila H, Castrén E (2004) Effects of NMDA-receptor antagonist treatment on c-fos expression in rat brain areas implicated in schizophrenia. Cell Mol Neurobiol 24:769–780

    Article  PubMed  Google Scholar 

  • van den Buuse M (2010) Modeling the positive symptoms of schizophrenia in genetically modified mice: pharmacology and methodology aspects. Schizophr Bull 36:246–270

    Article  PubMed Central  PubMed  Google Scholar 

  • Vijayraghavan S, Wang M, Birnbaum SG, Williams GV, Arnsten AF (2007) Inverted-U dopamine D1 receptor actions on prefrontal neurons engaged in working memory. Nat Neurosci 10:376–384

    Article  CAS  PubMed  Google Scholar 

  • Williams GV, Goldman-Rakic PS (1995) Modulation of memory fields by dopamine D1 receptors in prefrontal cortex. Nature 376:572–575

    Article  CAS  PubMed  Google Scholar 

  • Xu X, Domino EF (1994) Genetic differences in the locomotor response to single and daily doses of phencyclidine in inbred mouse strains. Behav Pharmacol 5:623–629

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

This work has received support from the Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM (13INT4 intramural project), and the Innovative Medicine Initiative Joint Undertaking under grant agreement no. 115008 of which resources are composed of EFPIA in-kind contribution and financial contribution from the European Union’s Seventh Framework Programme (FP7/2007-2013). We thank Emilio Regli, Patricia Sariñana, and Miguel Angel López-Venegas, as well as Leticia Campa, Mireia Galofré, Noemí Jurado, and Verónica Paz for technical support.

Conflict of interest

Francesc Artigas has received consulting and educational honoraria from Lundbeck, and he is PI of a grant from Lundbeck. He is also member of the scientific advisory board of Neurolixis. The rest of authors declare no conflict of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Anna Castañé.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Castañé, A., Santana, N. & Artigas, F. PCP-based mice models of schizophrenia: differential behavioral, neurochemical and cellular effects of acute and subchronic treatments. Psychopharmacology 232, 4085–4097 (2015). https://doi.org/10.1007/s00213-015-3946-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00213-015-3946-6

Keywords

Navigation