You are on page 1of 20

HHS Public Access

Author manuscript
Int J Neuropsychopharmacol. Author manuscript; available in PMC 2016 April 19.
Author Manuscript

Published in final edited form as:


Int J Neuropsychopharmacol. 2013 November ; 16(10): 2131–2144. doi:10.1017/S1461145713000606.

Preclinical models of antipsychotic drug action


José L. Moreno1 and Javier González-Maeso1,2,3
1Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
2Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, USA
3Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
Author Manuscript

Abstract
One of the main obstacles faced by translational neuroscience is the development of animal
models of psychiatric disorders. Behavioural pharmacology studies indicate that psychedelic
drugs, such as lysergic acid diethylamide (LSD) and dissociative drugs, such as phencyclidine
(PCP), induce in healthy human volunteers psychotic and cognitive symptoms that resemble some
of those observed in schizophrenia patients. Serotonin 5-HT2A and metabotropic glutamate 2
receptors have been involved in the mechanism of action of psychedelic and dissociative drugs.
Here we review recent advances using LSD-like and PCP-like drugs in rodent models that
implicate these receptors in the neurobiology of schizophrenia and its treatment.

Keywords
Author Manuscript

Antipsychotic drugs; epigenetics; lysergic acid diethylamide; metabotropic glutamate 2 receptor;


mouse models; phencyclidine; schizophrenia; serotonin 5-HT2A receptor

Introduction
Schizophrenia is a chronic mental disorder that afflicts nearly 1% of the population
worldwide (Sawa and Snyder, 2002; Freedman, 2003; Tamminga and Holcomb, 2005; Lewis
and Gonzalez-Burgos, 2006; Ross et al., 2006; van Os and Kapur, 2009; Abbott, 2010;
Dobbs, 2010). The first symptoms of schizophrenia generally occur in late adolescence or
early adulthood and continue into adult life. These include positive symptoms, such as
hallucinations and delusions, and negative symptoms such as alogia (poverty of speech),
avolition (lack of motivation) and anhedonia (lack of pleasure). Cognitive deficits, including
Author Manuscript

altered working memory and disordered thought, are present in the majority of
schizophrenia patients and are responsible for a significant portion of impairments and
disabilities associated with schizophrenic disorders (Green, 1996; Barch and Ceaser, 2012;
Minzenberg and Carter, 2012; Curley et al., 2013). It has also been shown that suicide is one
the principal causes of death in schizophrenia patients, with a 50% lifetime risk for suicide

Address for correspondence: Dr J. González-Maeso, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One
Gustave L. Levy Place, Box 1229, New York, NY 10029, USA. Tel.: 212 824 8973 Fax: 646 537 9583, Javier.Maeso@mssm.edu.
Statement of Interest
None.
Moreno and González-Maeso Page 2

attempts in patients with schizophrenia as compared with approximately 5% in the general


Author Manuscript

population (Meltzer et al., 2003a; Hawton et al., 2005; Pompili et al., 2007; Saha et al.,
2007).

It is well established that genetics plays a significant role in schizophrenia and schizophrenia
spectrum disorders (International Schizophrenia Consortium, 2008; Stefansson et al., 2008,
2009; Walsh et al., 2008; Purcell et al., 2009; Vacic et al., 2011). However, the study of
family members supports the conclusion that genetic factors are not sufficient to cause
schizophrenia (Tsuang et al., 2001). As discussed earlier, the lifetime prevalence of
schizophrenia in the general population is about 1%. This risk of developing schizophrenia
increases with the degree of genetic relatedness to the patient. As an example, third-degree
relatives (e.g. first cousins) share about 12.5% of their genes and show a risk of developing
schizophrenia of approximately 2%. Second-degree relatives (e.g. half-siblings) share
approximately 25% of the genes and show a risk of 6%; whereas first-degree relatives (e.g.
Author Manuscript

siblings and dizygotic twins) share approximately 50% of the genes and show a risk of about
9%. Monozygotic twins, whose DNA sequences are almost 100% identical, have a
concordance for schizophrenia of nearly 50% (McGuffin et al., 1994; Cardno and
Gottesman, 2000; Gottesman and Erlenmeyer-Kimling, 2001). Such results support the
conclusion that genetics plays a significant role in the aetiology of schizophrenia. At the
same time, they also favour a significant contribution of environmental factors in the
development of this complex disease (Lewis and Levitt, 2002; Bale et al., 2010).
Epidemiological studies have demonstrated that maternal adverse life-events during
pregnancy such as famine, war and death of a relative, increase the risk of schizophrenia in
the new-borns (Bradley and Dinan, 2010; Markham and Koenig, 2011). It has also been
shown that maternal infection during pregnancy raises the risk for schizophrenia in the
offspring (Yolken and Torrey, 2008; Brown and Derkits, 2010; Patterson, 2011). Of
Author Manuscript

particular interest and one of the focuses of this review is the generation of animal models
that may help understand the molecular mechanisms through which maternal/foetal
gene×environment interactions contribute to the onset of schizophrenia and other psychiatric
disorders later in life.

The antipsychotic drugs currently available for clinical use are able to ameliorate some of
the symptoms in schizophrenia patients, rather than to reverse the underlying alterations
responsible for the disease. Because of this, schizophrenia is still defined as an incurable
psychiatric disorder and its treatment typically continues for life. It is reasonable to suspect
that schizophrenia, as currently defined, represents a unique human disorder and,
consequently, modelling its phenotypic expression in rodent models remains controversial
(Arguello and Gogos, 2006; Powell and Miyakawa, 2006; Powell and Geyer, 2007; Geyer,
Author Manuscript

2008; Pratt et al., 2008; Kellendonk et al., 2009; O’Tuathaigh et al., 2009; Nestler and
Hyman, 2010; van den Buuse, 2010; Fernando and Robbins, 2011; Geyer et al., 2012). The
neurodevelopmental stages, as well as the anatomical location and neuronal circuits involved
in processes of cognition and perception, differ between humans and rodents, which also
raises concerns with respect to the translation of preclinical findings into clinical use.
Consequently, one of the limitations in molecular psychiatry in general and in schizophrenia
research in particular, is the development of rodent models that may help identify new
targets for antipsychotic drug development. In this review, we summarize recent advances in

Int J Neuropsychopharmacol. Author manuscript; available in PMC 2016 April 19.


Moreno and González-Maeso Page 3

our understanding, based on rodent models of psychedelic or dissociative drug exposure, of


Author Manuscript

not only the complex mechanisms associated with antipsychotic drug action, but also the
origins of schizophrenia itself.

Pharmacological models of psychosis and antipsychotic drug action


Glutamatergic dissociative drugs such as phencyclidine (PCP), ketamine and dizolcipine
(MK801), behave as non-competitive N-methyl-D-aspartate receptor antagonists (Morris et
al., 2005; Kristiansen et al., 2007). In an innovative study, Krystal et al. (1994) demonstrated
that administration of sub-anaesthetic doses of ketamine into healthy volunteers induces
negative symptoms and alterations in working memory and cognition that resemble those
observed in schizophrenia patients. A single administration of ketamine does not recreate
positive symptoms such as hallucinations, although paranoia and thought disorder are
induced (Krystal et al., 1994). Follow-up studies testing the effects of dissociative drugs in
Author Manuscript

healthy volunteers further validated these findings, suggesting alterations in time and
sensory perception, thought, speech, mood and affect that resemble some of the psychotic
states, negative symptoms and cognitive deficits associated with schizophrenia
(Vollenweider et al., 1997a, b, 2000; Adler et al., 1999; Newcomer et al., 1999; Umbricht et
al., 2000, 2002; Cho et al., 2005; Krystal et al., 2005; Schmidt et al., 2012, 2013; Driesen et
al., 2013). This is supported by the effects of chronic abuse of PCP, which commonly lead to
a misdiagnosis of schizophrenia (Rainey and Crowder, 1975; Allen and Young, 1978;
Pearlson, 1981; Cosgrove and Newell, 1991), whereas PCP administration to schizophrenia
patients who are not receiving antipsychotic treatment aggravates positive symptoms (Lahti
et al., 2001). Together, these findings point toward a potential dysregulation in glutamatergic
synaptic transmission as potentially involved in negative and cognitive symptoms of
schizophrenia, as well as in positive symptoms, and support the use of PCP-like dissociative
Author Manuscript

drugs in animals as a preclinical model of psychosis-like behaviour.

Serotonin (5-HT), and in particular the 5-HT2A receptor, has received much attention with
regard to schizophrenia and psychosis (Geyer and Vollenweider, 2008; Aghajanian, 2009;
Gonzalez-Maeso and Sealfon, 2009b; Geyer et al., 2012). The hallucinogenic properties of
the semi-synthetic compound d-lysergic acid diethylamide (LSD) were discovered
serendipitously by Albert Hoffman in 1943 (Hofmann, 1959). The 5-HT hypothesis about
the mechanism of action of LSD was proposed based on the structural similarities between
5-HT and hallucinogenic drugs such as LSD and psilocin, and the identification of 5-HT as a
neurotransmitter (Dahlstrom and Fuxe, 1964). Later in the 1980s, Richard Glennon, Milt
Titeler and their collaborators showed a highly significant correlation between the affinities
of 22 hallucinogenic compounds for 5-HT2 binding sites and their hallucinogenic potency in
Author Manuscript

humans (Glennon et al., 1984). This milestone work also demonstrated a significant
correlation between the 5-HT2 binding affinities of these agents and drug discrimination
ED50 values in rats (Glennon et al., 1984). Further work in healthy volunteers showed that
the psychoactive effects of psychedelic drugs such as LSD, mescaline, psilocybin, and N, N-
dimethyltryptamine drugs share several features with schizophrenia, including: perceptual
disturbances; sensory processing; cognition; changes in brain metabolism; self-
representation (Young, 1974; Hermle et al., 1992; Vollenweider et al., 1998; Gouzoulis-
Mayfrank et al., 2005; Quednow et al., 2011; Carhart-Harris et al., 2012). Indeed, LSD

Int J Neuropsychopharmacol. Author manuscript; available in PMC 2016 April 19.


Moreno and González-Maeso Page 4

effects were one of the earliest models of schizophrenia (Keeler, 1965) and are currently
Author Manuscript

believed to model in normal subjects positive symptoms (e.g. hallucinations and delusions)
comparable to those seen in drug-naive first-episode schizophrenia patients (Geyer and
Vollenweider, 2008; Gonzalez-Maeso and Sealfon, 2009b; Geyer et al., 2012) through a
mechanism that involves activation of the 5-HT2A receptor in cortical glutamatergic neurons
(Beique et al., 2007; Gonzalez-Maeso et al., 2007; Celada et al., 2008). It has also been
suggested that relatives of schizophrenic patients are more susceptible to the psychotic
responses induced by LSD (Anastasopoulos and Photiades, 1962). Although LSD-like drugs
as a model of psychosis have limitations (Hays and Tilley, 1973), these findings suggest that
a better understanding of their molecular mechanism of action may provide additional
insights into the brain processes underlying non-drug-induced psychoses.

Given that schizophrenia is exclusively human, the generation of animal models remains
controversial. However, based on their effects in healthy volunteers, psychedelic and
Author Manuscript

dissociative drugs have been considered effective as a tool to model distinct aspects of
schizophrenia in rodents and consequently disruption of their behavioural effects has been
used as a model that predicts antipsychotic responses in terms of positive, negative or
cognitive deficits, depending on the behavioural paradigm used, as well as on whether PCP-
like or LSD-like drugs are employed to model psychosis. Some of these behavioural models
include those following.

Modulation of locomotor activity


In rodents, PCP-like drugs induce hyperlocomotor activity and modulation of this
behavioural effect has been repeatedly used as a model of antipsychotic action. It has been
suggested that the 5-HT2A receptor is at least in part involved in the effects of atypical (e.g.
clozapine, risperidone and olanzapine), but not typical (e.g. haloperidol and
Author Manuscript

chlorpromazine), antipsychotics on the hyperlocomotor response induced by dissociative


drugs (Gleason and Shannon, 1997; Bradford et al., 2010; Fribourg et al., 2011; Yadav et al.,
2011b). The authors demonstrated that the locomotor response induced by PCP-like drugs in
mice was reduced by haloperidol only at doses at which it also reduces the locomotor
response in the absence of PCP-like drugs. In contrast, the atypical antipsychotics clozapine
and olanzapine reduced the locomotor activity induced by PCP-like drugs at doses that were
significantly lower than those that alone affect exploratory behaviour. Atypical
antipsychotics all have in common a high affinity for the 5-HT2A receptor and a much lower
affinity for the dopamine D2 receptor (Miyamoto et al., 2005; Lieberman et al., 2008).
Typical antipsychotics, on the contrary, have an antagonistic effect at the dopamine D2
receptor that has been involved in their mechanism of action (Carlsson et al., 2001; Meltzer
et al., 2003b). Importantly, it has been demonstrated that selective 5-HT2A receptor
Author Manuscript

antagonists such as volinanserin (M100907) reduce the locomotor hyperactivity induced by


PCP-like drugs in rats and that these doses by themselves do not affect exploratory
behaviour (Maurel-Remy et al., 1995). Similar data were obtained in mice (Carlsson, 1995).
The role of 5-HT2A receptors in the effects of clozapine on the locomotor response induced
by PCP-like drugs has been recently supported by the use of 5-HT2A knockout mice
(Fribourg et al., 2011). Although the pre-synaptic component of the serotonergic system
plays an important role in the antipsychotic-like properties of clozapine (Yadav et al.,

Int J Neuropsychopharmacol. Author manuscript; available in PMC 2016 April 19.


Moreno and González-Maeso Page 5

2011a), these results with knockout mice suggest that the 5-HT2A receptor is at least
Author Manuscript

involved in the therapeutic-like effects of atypical antipsychotics, using hyperlocomotor


activity induced by PCP-like drugs as a rodent model of psychosis. Recent findings also
point toward a role of 5-HT2A receptors expressed in cortical glutamatergic neurons in the
effects of high doses of clozapine, but not haloperidol or risperidone, on locomotor
suppression (McOmish et al., 2012; Williams et al., 2012).

The psychedelic 5-HT2A/2C receptor agonist 2,5-dimethoxy-4-iodoamphetamine (DOI) has


been shown to affect locomotor activity in mice (Gonzalez-Maeso et al., 2007; Halberstadt
et al., 2009, 2013) and rats (Krebs-Thomson et al., 1998; Van Oekelen et al., 2003; Marona-
Lewicka et al., 2005). Low doses of DOI significantly increase locomotor activity, an effect
that is absent in 5-HT2A knockout mice (Halberstadt et al., 2009). However, locomotor
activity and exploratory behaviour is reduced in mice injected with higher doses of DOI, an
effect that remains unaffected in 5-HT2A knockout mice and is reversed by the selective 5-
Author Manuscript

HT2C receptor antagonist SER-082 (Halberstadt et al., 2009). These results suggest that both
5-HT2A and 5-HT2C receptors are involved in the inverted U-shaped dose–response effect of
DOI on the locomotor response in mice. A more detailed characterization of the effects of
phenylalkylamine psychedelics, including low doses of DOI, mescaline, 2,5-dimethoxy-4-
ethylamphetamine, 2,5-dimethoxy-4-propylamphetamine, 2,4,5-trimethoxyamphetamine
and 4-bromo-3,6-dimethoxybenzocyclobuten-1-yl-methylamine (TCB-2), shows increased
locomotor activity in wild-type mice, an effect that is blocked by M100907 and abolished in
5-HT2A knockout mice (Halberstadt et al., 2013). Interestingly, the effect on locomotor
hyperactivity induced by phenylalkylamine psychedelics is not induced by the non-
psychedelic phenylalkylamine 2,5-dimethoxy-4-tert-butylamphetamine (DOTB; Halberstadt
et al., 2013). These findings suggest that activation of the 5-HT2A receptor by psychedelics,
but not by non-psychedelics, increases locomotor activity in mice, which may serve as a
Author Manuscript

model of hallucinogenic potential in humans. Using pharmacological tools in rats, it has


been shown that the effects of the phenylalkylamine DOI on exploratory behaviour are
reversed by the 5-HT2A receptor antagonist M100907, whereas those of the ergoline LSD
are not (Krebs- Thomson et al., 1998). Together, these findings in mice and rats support the
link between activation of the 5-HT2A receptor and the unique behavioural effects of
phenylalkylamine psychedelics. However, further work is needed to define the
pharmacological and molecular mechanisms, such as activation of dopamine D2 receptors
(Marona-Lewicka et al., 2005) underlying the differential effects of phenylalkylamine and
ergoline psychedelics on locomotor activity and other rodent behaviours.

Head-twitch behaviour
Head-twitch behavioural response in rodents is a rapid lateral movement of the head similar
Author Manuscript

to the pinna reflex (Canal and Morgan, 2012). It has been shown to be induced by a variety
of psychedelic 5-HT2A receptor agonists such as DOI, 1-(2,5-dimethoxy-4-methylphenyl)-2-
aminopropane, 1-(2,5-dimethoxy-4-bromophenyl)-2-aminopropane (DOB), mescaline, LSD,
TCB-2 and psilocin (Gonzalez-Maeso et al., 2003, 2007; Benneyworth et al., 2008; Canal et
al., 2010; Fox et al., 2010; Halberstadt and Geyer, 2013) and reversed by 5-HT2A receptor
antagonists (Fantegrossi et al., 2005; Benneyworth et al., 2008; Fribourg et al., 2011). This
behaviour is different as compared to head-weaving (slow, side-to-side lateral head

Int J Neuropsychopharmacol. Author manuscript; available in PMC 2016 April 19.


Moreno and González-Maeso Page 6

movements induced by dissociative drugs; Kozlenkov and Gonzalez-Maeso, 2012) and wet-
Author Manuscript

dog shake behaviour (repetitive shaking of the body commonly observed in rodents during
morphine withdrawal; Martin et al., 1963). Importantly, the head-twitch behaviour induced
by psychedelic drugs is absent in 5-HT2A knockout mice (Gonzalez-Maeso et al., 2003) and
rescued in mice that express 5-HT2A receptors only in cortical glutamatergic neurons
(Gonzalez-Maeso et al., 2007). Direct injection of psychedelic drugs into medial prefrontal
cortex provides additional evidence that head-twitch behaviour is mediated through cortical
5-HT2A receptors (Willins and Meltzer, 1997; see also Gresch et al., 2002 for data
suggesting a role of cortical 5-HT2A receptors in the discriminative stimulus properties of
LSD).

A long-lasting question in neuropharmacology has been the molecular mechanisms


underlying the unique behavioural responses induced by psychedelics as compared with
those induced by non-psychedelic 5-HT2A receptor agonists (Nichols, 2004; Gonzalez-
Author Manuscript

Maeso and Sealfon, 2009a). Thus, non-psychedelic 5-HT2A receptor agonists such as
lisuride, ergotamine and DOTB, bind with high affinity to and activate the 5-HT2A receptor
with similar potencies and efficacies as those observed by psychedelic drugs (Egan et al.,
1998; Hoyer et al., 2002). It has been recently shown that only 5-HT2A receptor agonists
with psychedelic potential in humans induce a significant head-twitch behaviour in mice and
that this behavioural response is not induced by non-psychedelic drugs such as lisuride and
ergotamine (Gonzalez-Maeso et al., 2003, 2007). Similar results have recently been reported
with the use of a head-mounted magnet and a magnetometer coil (Halberstadt and Geyer,
2013). Together, these findings suggest that head-twitch behaviour may be used as a rodent
behavioural model of psychedelic potential in humans.

Modulation of psychedelic-dependent head-twitch behaviour has also been tested as a model


Author Manuscript

of antipsychotic action (see later for discussion).

Prepulse inhibition of startle


The development of psychosis is the most striking element of schizophrenia, but deficits in
the sensory gating have now been shown as core features of the illness (Barch and Ceaser,
2012; Lewis et al., 2012; Minzenberg and Carter, 2012). Prepulse inhibition (PPI) is a
behavioural model that measures sensorimotor gating. In this model, a weak pre-stimulus
(auditory or tactile) reduces the startle response to a subsequent intense startling stimulus
(Geyer and Ellenbroek, 2003). Patients with schizophrenia exhibit deficits in PPI models of
the acoustic startle reflex (Ludewig et al., 2003) and impaired PPI response can be used as a
measure of alterations in sensorimotor gating in healthy volunteers (Heekeren et al., 2007).
PCP-like drugs produce PPI deficits in mice (Dulawa and Geyer, 1996), rats (Mansbach and
Author Manuscript

Geyer, 1989, 1991) and non-human primates (Javitt and Lindsley, 2001; Linn and Javitt,
2001). It has also been demonstrated that the psychedelic drugs DOI and LSD disrupt PPI,
an effect that is reversed by selective 5-HT2A antagonists such as M100907 (Sipes and
Geyer, 1997; Ouagazzal et al., 2001; McFarland et al., 2011). However, compounds that lack
psychedelic action (such as lisuride and yohimbine) also disrupt PPI of the acoustic startle in
rodent models (Bell et al., 2003; Swerdlow et al., 2003; Powell et al., 2005; Halberstadt and
Geyer, 2010). Although this raises concerns about the validity of assessing deficits in PPI as

Int J Neuropsychopharmacol. Author manuscript; available in PMC 2016 April 19.


Moreno and González-Maeso Page 7

a rodent behaviour model that predicts psychedelic activity, the possibility of cross-species
Author Manuscript

measurements of sensorimotor gating supports the use of PPI of startle in the identification
of new antipsychotic medications with special emphasis on attentional deficits present in
schizophrenia patients.

Behaviour models of metabotropic glutamate 2-dependent antipsychotic


action
All antipsychotic drugs currently used in the clinic are dopaminergic and serotonergic
receptor ligands. Relatively recent findings using dissociative (Moghaddam and Adams,
1998) and psychedelic (Gewirtz and Marek, 2000) drugs in preclinical models of
antipsychotic responses led to the first clinical studies with the metabotropic glutamate 2 and
3 (mGlu2/3) receptor orthosteric agonist LY404039 (active compound of LY2140023; Patil et
Author Manuscript

al., 2007; orthosteric agonists bind to the same binding site as the endogenous
neurotransmitter glutamate). These data suggested that LY2140023 represents a potential
new approach to treat schizophrenia. However, in two follow-up studies, Eli Lilly and
Company published double-blind phase 2 clinical trials showing that neither LY2140023 nor
olanzapine were more efficacious than placebo (Kinon et al., 2011), which are inconclusive
findings, and that LY2140023 does not separate from placebo whereas the positive control
risperidone was efficacious (Eli Lilly and Company, 2012a). These findings preceded the
press release that announced the decision to stop the on-going phase 3 clinical trial of the
orthosteric mGlu2/3 agonist for the treatment of schizophrenia (Eli Lilly and Company,
2012b). These discouraging findings contrast recent publications with the allosteric positive
modulator of the mGlu2 receptor ADX71149 from Addex Inc. in partnership with Janssen
R&D (Addex Therapeutics, 2012). Allosteric positive modulators bind to a site distinct from
that of the endogenous neurotransmitter; they do not affect basal receptor activity but
Author Manuscript

potentiate the effect of orthosteric agonists. The clinical data show safety and tolerability
and demonstrate an effect in negative symptoms of schizophrenia patients (Addex
Therapeutics, 2012). Although promising, more molecular, preclinical and clinical data are
needed to validate whether activation of the mGlu2 receptor may serve as a new approach to
treat this psychiatric disease.

Another question of interest is the therapeutic-like efficacy of different orthosteric mGlu2/3


agonists and allosteric mGlu2 receptor agonists. Most of the preclinical studies assessing the
role of mGlu2 and/or mGlu3 receptors in the antipsychotic-like effects of mGlu2/3 receptor
agonists have been conducted with orthosteric mGlu2/3 receptor agonists such as LY379268
and LY354740 (Gewirtz and Marek, 2000; Klodzinska et al., 2002; Linden et al., 2004;
Winter et al., 2004; Gonzalez-Maeso et al., 2008; Woolley et al., 2008; Uslaner et al., 2009;
Author Manuscript

Horiguchi et al., 2011; Jones et al., 2011; Moreno et al., 2011b). At present, there is only one
study describing the antipsychotic-like effects of the orthosteric mGlu2/3 receptor agonist
LY404039 (active compound of its prodrug LY2140023 in clinical trials; Fell et al., 2008).
The authors tested the effects of LY404039 on the hyperlocomotor activity induced by PCP
and amphetamine in mice. While there are limits to the extrapolation of these findings to
humans, only a relatively high dose of LY404039 (10 mg/kg) significantly reduced the PCP-
induced locomotor activity in wild-type mice (Fell et al., 2008). This antipsychotic-like

Int J Neuropsychopharmacol. Author manuscript; available in PMC 2016 April 19.


Moreno and González-Maeso Page 8

effect was absent in mGlu2 knockout, but not in mGlu3 knockout, mice. Similar mGlu2-
Author Manuscript

dependent effects have been reported with the mGlu2/3 receptor agonist LY379268 (Woolley
et al., 2008). Thus, the effect of LY379268 on the PCP-induced locomotor activity was
observed in wild-type and mGlu3 knockout, but not mGlu2 knockout mice (Woolley et al.,
2008). However, and importantly, the antipsychotic-like effect of LY379268 on PCP-induced
behaviour was seen at doses below 1 mg/kg (Woolley et al., 2008). Consistent with the
hypothesis that mGlu2, and not mGlu3, is necessary for the antipsychotic-like effects of
mGlu2/3 agonists, data with the allosteric mGlu2 agonists biphenyl-indanone A
(Benneyworth et al., 2007) and LY487379 (Galici et al., 2005) show antipsychotic-like
behavioural effects using the psychedelic drug DOB (Benneyworth et al., 2007), the
dissociative drug PCP (Galici et al., 2005) and amphetamine (Galici et al., 2005) as rodent
models of psychosis. Further clinical studies are needed to evaluate the true efficacy and
long-term safety and these agents in schizophrenia patients.
Author Manuscript

With regard to the molecular mechanism of action of glutamate antipsychotics, previous


findings suggest that a functional crosstalk between mGlu2 and 5-HT2A receptors is
necessary for the antipsychotic-like effects of LY379268 (Gonzalez-Maeso et al., 2008;
Fribourg et al., 2011; Moreno et al., 2011a, 2012). Thus, it was shown that the antipsychotic-
like effect of LY379268 (5 mg/kg) on the hyperlocomotor behaviour induced by the PCP-
like drug MK801 was mGlu2-dependent and absent in 5-HT2A knockout mice (Fribourg et
al., 2011). The authors also demonstrate that some, but not all, mGlu2/3 receptor agonists
bind mGlu2 receptor in complex with the 5-HT2A receptor and alter the pattern of
heterotrimeric G protein coupling in a way that predicts their antipsychotic-like behavioural
effects (Fribourg et al., 2011). This represents the first biochemical assay that predicts
psychoactive behavioural effects of a variety of serotonergic and glutamatergic compounds
(Fribourg et al., 2011). Whether the antipsychotic-like effects of LY404039 and other
Author Manuscript

orthosteric mGlu2/3, and allosteric mGlu2 receptor agonists require expression of the 5-
HT2A receptor remains to be investigated. Nevertheless, examples of non-antipsychotic
drugs that induce antipsychotic-like behaviour in rodents include methysergide, mianserin
and ketanserin (Ninan and Kulkarni, 1998; O’Neill et al., 1998; Fletcher et al., 2011;
Fribourg et al., 2011; Yadav et al., 2011b; Mestre et al., 2013).

Behaviour models of chronic antipsychotic treatment


In schizophrenia patients, antipsychotic drugs are administered chronically (weeks to
months of continuous treatment; Agid et al., 2003). However, most of the animal behaviour
models of antipsychotic action are based on a single administration of the drug to be tested
(Geyer et al., 2012), which raises concerns about the validity of this therapeutic-like
Author Manuscript

behaviour. Recent findings have shown that down-regulation of 5-HT2A receptor may be one
of the mechanisms underlying the antipsychotic-like effects induced by chronic treatment
with antipsychotic drugs (Yadav et al., 2011b). The authors assessed the effects of chronic
treatment with antipsychotic drugs (clozapine, olanzapine and haloperidol) and with 5-HT2A
receptor antagonists/inverse agonists (ketanserin, M100907, M11939, SR46349B and
primavanserin) on 5-HT2A receptor protein level and PCP-induced hyperlocomotor activity
(Yadav et al., 2011b). Interestingly, chronic treatment with clozapine and olanzapine, but not
haloperidol, induced down-regulation of 5-HT2A receptor protein in mouse frontal cortex

Int J Neuropsychopharmacol. Author manuscript; available in PMC 2016 April 19.


Moreno and González-Maeso Page 9

and decreased the psychosis-like effects of PCP. These effects were not observed after
Author Manuscript

chronic treatment with M100907, M11939, SR46349B or primavanserin. Chronic treatment


with the non-antipsychotic ketanserin down-regulated 5-HT2A receptor protein, whereas
PCP induced hyperlocomotion was not affected (Yadav et al., 2011b).

Another approach to determine the therapeutic-like effects of chronic treatment with


antipsychotic drugs is based on the use of LSD as a mouse model of psychosis (Gonzalez-
Maeso et al., 2008; Kurita et al., 2012; Moreno et al., 2013a, b). Administration of
psychedelic 5-HT2A receptor agonists induces expression of the immediate early genes c-
fos, egr-1 and egr-2 in mouse somatosensory cortex (Gonzalez-Maeso et al., 2003,2007;
Moreno et al., 2013a, b). The authors found that chronic (21 d), but not sub-chronic (2 d),
treatment with clozapine eliminates the induction of c-fos, egr-1 and egr-2 by LSD 1 d after
the final injection (Moreno et al., 2013b). The induction of c-fos, egr-1 and egr-2 by LSD
was not affected with chronic treatment with haloperidol. These findings correlate with the
Author Manuscript

effect of chronic clozapine, but not chronic haloperidol, on 5-HT2A receptor binding in
mouse somatosensory cortex. Thus, chronic, but not sub-chronic, treatment with clozapine
down-regulated [3H]ketanserin binding, an effect that was not observed after chronic
treatment with haloperidol (Moreno et al., 2013b). These findings suggest that down-
regulation of 5-HT2A receptor is one of the mechanisms underlying the therapeutic effects of
chronic treatment with antipsychotic drugs. Further work is needed to understand the
cellular networks responsible for down-regulation of this receptor with antagonists/inverse
agonists such as clozapine and olanzapine, as well as their sedative effects (McOmish et al.,
2012; Williams et al., 2012).

Rodent models of maternal adverse life-events during pregnancy


Author Manuscript

While it is clear that genetics plays an important role in schizophrenia (Stefansson et al.,
2008, 2009; Walsh et al., 2008; Purcell et al., 2009; Vacic et al., 2011), the genetic
mechanisms of transmission are complex as demonstrated by studies of familial segregations
and monozygotic twins. Thus, twin studies have shown that the risk for both identical twins
to develop schizophrenia is 30–50% (see earlier). Such results point toward a significant
contribution of environmental factors in the development of this neurodevelopmental
disease. Interestingly, epidemiological studies repeatedly suggest that maternal
environmental factors during pregnancy, such as pathogen infection (e.g. virus, bacteria and
protozoa; Menninger, 1919; Brown et al., 2001, 2004, 2005; Babulas et al., 2006; Sorensen
et al., 2009; Yudofsky, 2009) and adverse life-events such as famine (Susser et al., 2008),
war (van Os and Selten, 1998; Malaspina et al., 2008) and death or illness in a first-degree
relative (Khashan et al., 2008) increase the risk of schizophrenia in the adult offspring. Also
Author Manuscript

shown has been an association between foetal hypoxia or anoxia due to obstetric
complications during labour and delivery and schizophrenia (Buka et al., 1993; Hirshfeld-
Becker et al., 2004; Mittal et al., 2008; Nicodemus et al., 2008). Interestingly, recent
findings demonstrate that mouse models of maternal viral infection and maternal variable
stress may serve as preclinical models for future studies to investigate the mechanisms and
alterations in gene×environment interactions responsible for schizophrenia symptoms.

Int J Neuropsychopharmacol. Author manuscript; available in PMC 2016 April 19.


Moreno and González-Maeso Page 10

Radioligand binding assays in post-mortem human brain samples suggest that 5-HT2A
Author Manuscript

receptor binding is increased and mGlu2/3 binding is decreased in frontal cortex of untreated
schizophrenic subjects (Gonzalez-Maeso et al., 2008; Moreno et al., 2012; Muguruza et al.,
2012). Similar alterations have been reported in two mouse models of maternal adverse life-
events: influenza virus infection (Moreno et al., 2011b); variable and unpredictable stress
(Holloway et al., 2013). Thus, adult mice born to influenza virus infected mothers show up-
regulation of 5-HT2A receptor and down-regulation of mGlu2/3 receptors (Moreno et al.,
2011b). These biochemical changes correlate with behavioural alterations: increased head-
twitch behaviour induced by the psychedelic drug DOI; decreased effect of LY379268 on
the MK801-dependent locomotor activity (Moreno et al., 2011b). Interestingly, a similar
pattern of biochemical and behavioural changes has been shown in adult mice born to
mothers stressed during pregnancy (Holloway et al., 2013). It has also been suggested that
the existence of epigenetic modifications at the promoter regions of 5-HT2A (Htr2a), mGlu2
Author Manuscript

(Grm2) and mGlu3 (Grm3) genes correlates with this pattern of expression and their
behavioural function (Matrisciano et al., 2012). As reviewed earlier, both maternal infection
and maternal severe stress during pregnancy are implicated as having an aetiological role in
schizophrenia. Numerous experimental investigations have provided robust evidence that
adverse life experiences profoundly modulate immune cell functions (Maes et al., 1998;
Steptoe et al., 2001; Vanbesien-Mailliot et al., 2007; Garcia-Bueno et al., 2008) and it is
thought that activation of the maternal immune system produces cytokines and chemokines
that ultimately alter neurogenesis and migration of neurons during brain development,
contributing to schizophrenia risk (Nawa and Takei, 2006; Bilbo and Schwarz, 2009;
Deverman and Patterson, 2009). Further investigation is necessary to understand the
gene×environment interactions as well as the contribution of specific components of the
maternal immune system responsible for schizophrenia and other neurodevelopmental
disorders in the adult offspring.
Author Manuscript

Conclusions
In this review, we summarize some of the current knowledge of animal models of psychosis
and antipsychotic action. The antipsychotics currently used in clinical practice are designed
to treat the symptoms of the disease. As yet, the underlying mechanisms responsible for the
onset of psychosis in schizophrenia patients remain largely unknown. About 30% of the
patients are considered treatment resistant and will continue to experience schizophrenia
symptoms despite the use of antipsychotic drug treatment. Recent preclinical (Kurita et al.,
2012) and clinical (Citrome et al., 2004; Meltzer et al., 2011) findings have proposed the use
of epigenetic drugs to inhibit histone deacetylation in order to improve the clinical efficacy
of the currently available antipsychotic drugs. Although psychedelic and dissociative drugs
Author Manuscript

as a model of schizophrenia have limitations, some of the findings summarized in this


review suggest that their use in animal models may serve as a tool to find new antipsychotic
drugs for treatment and also prevention of schizophrenia and other psychotic disorders.

Acknowledgements
This study was supported by NIH R01 MH084894, Dainippon Sumitomo Pharma, NARSAD and The Mortimer D.
Sackler Foundation (J.G.M.).

Int J Neuropsychopharmacol. Author manuscript; available in PMC 2016 April 19.


Moreno and González-Maeso Page 11

References
Author Manuscript

Abbott A. Schizophrenia: the drug deadlock. Nature. 2010; 468:158–159. [PubMed: 21068804]
Addex Therapeutics. 2012. Available at: http://www.addextherapeutics.com/investors/press-releases/
news-details/article/addex-reports-top-line-data-from-a-successful-phase-2a-clinical-study-with-
adx71149-in-schizophrenia/
Adler CM, Malhotra AK, Elman I, Goldberg T, Egan M, Pickar D, Breier A. Comparison of ketamine-
induced thought disorder in healthy volunteers and thought disorder in schizophrenia. Am J
Psychiatry. 1999; 156:1646–1649. [PubMed: 10518181]
Aghajanian GK. Modeling ‘psychosis’ in vitro by inducing disordered neuronal network activity in
cortical brain slices. Psychopharmacology (Berl). 2009; 206:575–585. [PubMed: 19241062]
Agid O, Kapur S, Arenovich T, Zipursky RB. Delayed-onset hypothesis of antipsychotic action: a
hypothesis tested and rejected. Arch Gen Psychiatry. 2003; 60:1228–1235. [PubMed: 14662555]
Allen RM, Young SJ. Phencyclidine-induced psychosis. Am J Psychiatry. 1978; 135:1081–1084.
[PubMed: 696930]
Anastasopoulos G, Photiades H. Effects of LSD-25 on relatives of schizophrenic patients. J Ment Sci.
Author Manuscript

1962; 108:95–98. [PubMed: 13861108]


Arguello PA, Gogos JA. Modeling madness in mice: one piece at a time. Neuron. 2006; 52:179–196.
[PubMed: 17015235]
Babulas V, Factor-Litvak P, Goetz R, Schaefer CA, Brown AS. Prenatal exposure to maternal genital
and reproductive infections and adult schizophrenia. Am J Psychiatry. 2006; 163:927–929.
[PubMed: 16648337]
Bale TL, Baram TZ, Brown AS, Goldstein JM, Insel TR, McCarthy MM, Nemeroff CB, Reyes TM,
Simerly RB, Susser ES, Nestler EJ. Early life programming and neurodevelopmental disorders.
Biol Psychiatry. 2010; 68:314–319. [PubMed: 20674602]
Barch DM, Ceaser A. Cognition in schizophrenia: core psychological and neural mechanisms. Trends
Cogn Sci. 2012; 16:27–34. [PubMed: 22169777]
Beique JC, Imad M, Mladenovic L, Gingrich JA, Andrade R. Mechanism of the 5-hydroxytryptamine
2A receptor-mediated facilitation of synaptic activity in prefrontal cortex. Proc Natl Acad Sci
USA. 2007; 104:9870–9875. [PubMed: 17535909]
Author Manuscript

Bell RL, Rodd ZA, Hsu CC, Lumeng L, Murphy JM, McBride WJ. Amphetamine-modified acoustic
startle responding and prepulse inhibition in adult and adolescent alcohol-preferring and -
nonpreferring rats. Pharmacol Biochem Behav. 2003; 75:163–171. [PubMed: 12759124]
Benneyworth MA, Xiang Z, Smith RL, Garcia EE, Conn PJ, Sanders-Bush E. A selective positive
allosteric modulator of metabotropic glutamate receptor subtype 2 blocks a hallucinogenic drug
model of psychosis. Mol Pharmacol. 2007; 72:477–484. [PubMed: 17526600]
Benneyworth MA, Smith RL, Sanders-Bush E. Chronic phenethylamine hallucinogen treatment alters
behavioral sensitivity to a metabotropic glutamate 2/3 receptor agonist.
Neuropsychopharmacology. 2008; 33:2206–2216. [PubMed: 17957214]
Bilbo SD, Schwarz JM. Early-life programming of later-life brain and behavior: a critical role for the
immune system. Front Behav Neurosci. 2009; 3:14. [PubMed: 19738918]
Bradford AM, Savage KM, Jones DN, Kalinichev M. Validation and pharmacological characterisation
of MK-801-induced locomotor hyperactivity in BALB/C mice as an assay for detection of novel
antipsychotics. Psychopharmacology (Berl). 2010; 212:155–170. [PubMed: 20676613]
Bradley AJ, Dinan TG. A systematic review of hypothalamic-pituitary-adrenal axis function in
Author Manuscript

schizophrenia: implications for mortality. J Psychopharmacol. 2010; 24:91–118. [PubMed:


20923924]
Brown AS, Derkits EJ. Prenatal infection and schizophrenia: a review of epidemiologic and
translational studies. Am J Psychiatry. 2010; 167:261–280. [PubMed: 20123911]
Brown AS, Cohen P, Harkavy-Friedman J, Babulas V, Malaspina D, Gorman JM, Susser ES, A.E.
Bennett Research Award. Prenatal rubella, premorbid abnormalities, and adult schizophrenia. Biol
Psychiatry. 2001; 49:473–486. [PubMed: 11257233]

Int J Neuropsychopharmacol. Author manuscript; available in PMC 2016 April 19.


Moreno and González-Maeso Page 12

Brown AS, Begg MD, Gravenstein S, Schaefer CA, Wyatt RJ, Bresnahan M, Babulas VP, Susser ES.
Serologic evidence of prenatal influenza in the etiology of schizophrenia. Arch Gen Psychiatry.
Author Manuscript

2004; 61:774–780. [PubMed: 15289276]


Brown AS, Schaefer CA, Quesenberry CP Jr. Liu L, Babulas VP, Susser ES. Maternal exposure to
toxoplasmosis and risk of schizophrenia in adult offspring. Am J Psychiatry. 2005; 162:767–773.
[PubMed: 15800151]
Buka SL, Tsuang MT, Lipsitt LP. Pregnancy/delivery complications and psychiatric diagnosis. A
prospective study. Arch Gen Psychiatry. 1993; 50:151–156. [PubMed: 8427556]
Canal CE, Morgan D. Head-twitch response in rodents induced by the hallucinogen 2, 5-dimethoxy-4-
iodoamphetamine: a comprehensive history, a re-evaluation of mechanisms, and its utility as a
model. Drug Test Anal. 2012; 4:556–576. [PubMed: 22517680]
Canal CE, Olaghere da Silva UB, Gresch PJ, Watt EE, Sanders-Bush E, Airey DC. The serotonin 2C
receptor potently modulates the head-twitch response in mice induced by a phenethylamine
hallucinogen. Psychopharmacology (Berl). 2010; 209:163–174. [PubMed: 20165943]
Cardno AG, Gottesman II. Twin studies of schizophrenia: from bow-and-arrow concordances to star
wars Mx and functional genomics. Am J Med Genet. 2000; 97:12–17. [PubMed: 10813800]
Author Manuscript

Carhart-Harris RL, Erritzoe D, Williams T, Stone JM, Reed LJ, Colasanti A, Tyacke RJ, Leech R,
Malizia AL, Murphy K, Hobden P, Evans J, Feilding A, Wise RG, Nutt DJ. Neural correlates of
the psychedelic state as determined by fMRI studies with psilocybin. Proc Natl Acad Sci USA.
2012; 109:2138–2143. [PubMed: 22308440]
Carlsson A, Waters N, Holm-Waters S, Tedroff J, Nilsson M, Carlsson ML. Interactions between
monoamines, glutamate, and GABA in schizophrenia: new evidence. Annu Rev Pharmacol
Toxicol. 2001; 41:237–260. [PubMed: 11264457]
Carlsson ML. The selective 5-HT2A receptor antagonist MDL 100,907 counteracts the psychomotor
stimulation ensuing manipulations with monoaminergic, glutamatergic or muscarinic
neurotransmission in the mouse–implications for psychosis. J Neural Transm. 1995; 100:225–237.
Celada P, Puig MV, Diaz-Mataix L, Artigas F. The hallucinogen DOI reduces low-frequency
oscillations in rat prefrontal cortex: reversal by antipsychotic drugs. Biol Psychiatry. 2008;
64:392–400. [PubMed: 18436196]
Cho HS, D’Souza DC, Gueorguieva R, Perry EB, Madonick S, Karper LP, Abi-Dargham A, Belger A,
Abi-Saab W, Lipschitz D, Bennet A, Seibyl JP, Krystal JH. Absence of behavioral sensitization in
Author Manuscript

healthy human subjects following repeated exposure to ketamine. Psychopharmacology (Berl).


2005; 179:136–143. [PubMed: 15682309]
Citrome L, Casey DE, Daniel DG, Wozniak P, Kochan LD, Tracy KA. Adjunctive divalproex and
hostility among patients with schizophrenia receiving olanzapine or risperidone. Psychiatr Serv.
2004; 55:290–294. [PubMed: 15001730]
Cosgrove J, Newell TG. Recovery of neuropsychological functions during reduction in use of
phencyclidine. J Clin Psychol. 1991; 47:159–169. [PubMed: 2026771]
Curley AA, Eggan SM, Lazarus MS, Huang ZJ, Volk DW, Lewis DA. Role of glutamic acid
decarboxylase 67 in regulating cortical parvalbumin and GABA membrane transporter 1
expression: implications for schizophrenia. Neurobiol Dis. 2013; 50:179–186. [PubMed:
23103418]
Dahlstrom A, Fuxe K. Localization of monoamines in the lower brain stem. Experientia. 1964;
20:398–399. [PubMed: 5856530]
Deverman BE, Patterson PH. Cytokines and CNS development. Neuron. 2009; 64:61–78. [PubMed:
Author Manuscript

19840550]
Dobbs D. Schizophrenia: the making of a troubled mind. Nature. 2010; 468:154–156. [PubMed:
21068803]
Driesen NR, McCarthy G, Bhagwagar Z, Bloch M, Calhoun V, D’Souza DC, Gueorguieva R, He G,
Ramachandran R, Suckow RF, Anticevic A, Morgan PT, Krystal JH. Relationship of resting brain
hyperconnectivity and schizophrenia-like symptoms produced by the NMDA receptor antagonist
ketamine in humans. Mol Psychiatry. 2013 Advance online publication. doi: 10.1038/mp.
2012.194.

Int J Neuropsychopharmacol. Author manuscript; available in PMC 2016 April 19.


Moreno and González-Maeso Page 13

Dulawa SC, Geyer MA. Psychopharmacology of prepulse inhibition in mice. Chin J Physiol. 1996;
39:139–146. [PubMed: 8955560]
Author Manuscript

Egan CT, Herrick-Davis K, Miller K, Glennon RA, Teitler M. Agonist activity of LSD and lisuride at
cloned 5HT2A and 5HT2C receptors. Psychopharmacology (Berl). 1998; 136:409–414. [PubMed:
9600588]
Eli Lilly and Company. 2012a. Available at: http://newsroom.lilly.com/releasedetail.cfm?
releaseid=690836
Eli Lilly and Company. 2012b. Available at: https://://investor.lilly.com/releaseDetail.cfm?
ReleaseID=703018
Fantegrossi WE, Harrington AW, Eckler JR, Arshad S, Rabin RA, Winter JC, Coop A, Rice KC,
Woods JH. Hallucinogen-like actions of 2,5-dimethoxy-4-(n)-propylthiophenethylamine (2C-T-7)
in mice and rats. Psychopharmacology (Berl). 2005; 181:496–503. [PubMed: 15983786]
Fell MJ, Svensson KA, Johnson BG, Schoepp DD. Evidence for the role of metabotropic glutamate
(mGlu)2 not mGlu3 receptors in the preclinical antipsychotic pharmacology of the mGlu2/3
receptor agonist (-)-(1R,4S,5S,6S)-4-amino-2-sulfonylbicyclo[3.1.0] hexane-4,6-dicarboxylic acid
(LY404039). J Pharmacol Exp Ther. 2008; 326:209–217. [PubMed: 18424625]
Author Manuscript

Fernando AB, Robbins TW. Animal models of neuropsychiatric disorders. Annu Rev Clin Psychol.
2011; 7:39–61. [PubMed: 21219191]
Fletcher PJ, Rizos Z, Noble K, Higgins Ga. Impulsive action induced by amphetamine, cocaine and
MK801 is reduced by 5-HT(2C) receptor stimulation and 5-HT(2A) receptor blockade.
Neuropharmacology. 2011; 61:468–477. [PubMed: 21402085]
Fox, Ma; French, HT.; LaPorte, JL.; Blackler, AR.; Murphy, DL. The serotonin 5-HT(2A) receptor
agonist TCB-2: a behavioral and neurophysiological analysis. Psychopharmacology. 2010;
212:13–23. [PubMed: 19823806]
Freedman R. Schizophrenia. N Engl J Med. 2003; 349:1738–1749. [PubMed: 14585943]
Fribourg M, et al. Decoding the signaling of a GPCR heteromeric complex reveals a unifying
mechanism of action of antipsychotic drugs. Cell. 2011; 147:1011–1023. [PubMed: 22118459]
Galici R, Echemendia NG, Rodriguez AL, Conn PJ. A selective allosteric potentiator of metabotropic
glutamate (mGlu) 2 receptors has effects similar to an orthosteric mGlu2/3 receptor agonist in
mouse models predictive of antipsychotic activity. J Pharmacol Exp Ther. 2005; 315:1181–1187.
Author Manuscript

[PubMed: 16123306]
Garcia-Bueno B, Caso JR, Leza JC. Stress as a neuroinflammatory condition in brain: damaging and
protective mechanisms. Neurosci Biobehav Rev. 2008; 32:1136–1151. [PubMed: 18468686]
Gewirtz JC, Marek GJ. Behavioral evidence for interactions between a hallucinogenic drug and group
II metabotropic glutamate receptors. Neuropsychopharmacology. 2000; 23:569–576. [PubMed:
11027922]
Geyer MA. Developing translational animal models for symptoms of schizophrenia or bipolar mania.
Neurotox Res. 2008; 14:71–78. [PubMed: 18790726]
Geyer MA, Ellenbroek B. Animal behavior models of the mechanisms underlying antipsychotic
atypicality. Prog Neuropsychopharmacol Biol Psychiatry. 2003; 27:1071–1079. [PubMed:
14642967]
Geyer MA, Vollenweider FX. Serotonin research: contributions to understanding psychoses. Trends
Pharmacol Sci. 2008; 29:445–453. [PubMed: 19086254]
Geyer MA, Olivier B, Joels M, Kahn RS. From antipsychotic to anti-schizophrenia drugs: role of
animal models. Trends Pharmacol Sci. 2012; 33:515–521. [PubMed: 22810174]
Author Manuscript

Gleason SD, Shannon HE. Blockade of phencyclidine-induced hyperlocomotion by olanzapine,


clozapine and serotonin receptor subtype selective antagonists in mice. Psychopharmacology
(Berl). 1997; 129:79–84. [PubMed: 9122367]
Glennon RA, Titeler M, McKenney JD. Evidence for 5-HT2 involvement in the mechanism of action
of hallucinogenic agents. Life Sci. 1984; 35:2505–2511. [PubMed: 6513725]
Gonzalez-Maeso J, Sealfon SC. Agonist-trafficking and hallucinogens. Curr Med Chem. 2009a;
16:1017–1027. [PubMed: 19275609]
Gonzalez-Maeso J, Sealfon SC. Psychedelics and schizophrenia. Trends Neurosci. 2009b; 32:225–232.
[PubMed: 19269047]

Int J Neuropsychopharmacol. Author manuscript; available in PMC 2016 April 19.


Moreno and González-Maeso Page 14

Gonzalez-Maeso J, Yuen T, Ebersole BJ, Wurmbach E, Lira A, Zhou M, Weisstaub N, Hen R,


Gingrich JA, Sealfon SC. Transcriptome fingerprints distinguish hallucinogenic and
Author Manuscript

nonhallucinogenic 5-hydroxytryptamine 2A receptor agonist effects in mouse somatosensory


cortex. J Neurosci. 2003; 23:8836–8843. [PubMed: 14523084]
Gonzalez-Maeso J, Weisstaub NV, Zhou M, Chan P, Ivic L, Ang R, Lira A, Bradley-Moore M, Ge Y,
Zhou Q, Sealfon SC, Gingrich JA. Hallucinogens recruit specific cortical 5-HT(2A) receptor-
mediated signaling pathways to affect behavior. Neuron. 2007; 53:439–452. [PubMed: 17270739]
Gonzalez-Maeso J, Ang RL, Yuen T, Chan P, Weisstaub NV, Lopez-Gimenez JF, Zhou M, Okawa Y,
Callado LF, Milligan G, Gingrich JA, Filizola M, Meana JJ, Sealfon SC. Identification of a
serotonin/glutamate receptor complex implicated in psychosis. Nature. 2008; 452:93–97.
[PubMed: 18297054]
Gottesman II, Erlenmeyer-Kimling L. Family and twin strategies as a head start in defining prodromes
and endophenotypes for hypothetical early-interventions in schizophrenia. Schizophr Res. 2001;
51:93–102. [PubMed: 11479071]
Gouzoulis-Mayfrank E, Heekeren K, Neukirch A, Stoll M, Stock C, Obradovic M, Kovar KA.
Psychological effects of (S)-ketamine and N,N-dimethyltryptamine (DMT): a double-blind, cross-
Author Manuscript

over study in healthy volunteers. Pharmacopsychiatry. 2005; 38:301–311. [PubMed: 16342002]


Green MF. What are the functional consequences of neurocognitive deficits in schizophrenia? Am J
Psychiatry. 1996; 153:321–330. [PubMed: 8610818]
Gresch PJ, Strickland LV, Sanders-Bush E. Lysergic acid diethylamide-induced Fos expression in rat
brain: role of serotonin-2A receptors. Neuroscience. 2002; 114:707–713. [PubMed: 12220572]
Halberstadt AL, Geyer MA. LSD but not lisuride disrupts prepulse inhibition in rats by activating the
5-HT(2A) receptor. Psychopharmacology. 2010; 208:179–189. [PubMed: 19937319]
Halberstadt AL, Geyer MA. Characterization of the head-twitch response induced by hallucinogens in
mice: detection of the behavior based on the dynamics of head movement. Psychopharmacology
(Berl). 2013; 227:727–739. [PubMed: 23407781]
Halberstadt AL, van der Heijden I, Ruderman MA, Risbrough VB, Gingrich JA, Geyer MA, Powell
SB. 5-HT(2A) and 5-HT(2C) receptors exert opposing effects on locomotor activity in mice.
Neuropsychopharmacology. 2009; 34:1958–1967. [PubMed: 19322172]
Halberstadt AL, Powell SB, Geyer MA. Role of the 5-HT2A receptor in the locomotor hyperactivity
produced by phenylalkylamine hallucinogens in mice. Neuropharmacology. 2013; 70C:218–227.
Author Manuscript

[PubMed: 23376711]
Hawton K, Sutton L, Haw C, Sinclair J, Deeks JJ. Schizophrenia and suicide: systematic review of risk
factors. Br J Psychiatry. 2005; 187:9–20. [PubMed: 15994566]
Hays P, Tilley JR. The differences between LSD psychosis and schizophrenia. Can Psychiatr Assoc J.
1973; 18:331–333. [PubMed: 4780752]
Heekeren K, Neukirch A, Daumann J, Stoll M, Obradovic M, Kovar KA, Geyer MA, Gouzoulis-
Mayfrank E. Prepulse inhibition of the startle reflex and its attentional modulation in the human S-
ketamine and N, N-dimethyltryptamine (DMT) models of psychosis. J Psychopharmacol. 2007;
21:312–320. [PubMed: 17591658]
Hermle L, Funfgeld M, Oepen G, Botsch H, Borchardt D, Gouzoulis E, Fehrenbach RA, Spitzer M.
Mescaline-induced psychopathological, neuropsychological, and neurometabolic effects in normal
subjects: experimental psychosis as a tool for psychiatric research. Biol Psychiatry. 1992; 32:976–
991. [PubMed: 1467389]
Hirshfeld-Becker DR, Biederman J, Faraone SV, Robin JA, Friedman D, Rosenthal JM, Rosenbaum
Author Manuscript

JF. Pregnancy complications associated with childhood anxiety disorders. Depress Anxiety. 2004;
19:152–162. [PubMed: 15129417]
Hofmann A. Psychotomimetic drugs, chemical and pharmacological aspects. Acta Physiol Pharmacol
Neerl. 1959; 8:240–258. [PubMed: 13852489]
Holloway T, Moreno JL, Umali A, Rayannavar V, Hodes GE, Russo SJ, Gonzalez-Maeso J. Prenatal
stress induces schizophrenia-like alterations of serotonin 2A and metabotropic glutamate 2
receptors in the adult offspring: role of maternal immune system. J Neurosci. 2013; 33:1088–1098.
[PubMed: 23325246]

Int J Neuropsychopharmacol. Author manuscript; available in PMC 2016 April 19.


Moreno and González-Maeso Page 15

Horiguchi M, Huang M, Meltzer HY. Interaction of mGlu2/3 agonism with clozapine and lurasidone to
restore novel object recognition in subchronic phencyclidine-treated rats. Psychopharmacology
Author Manuscript

(Berl). 2011; 217:13–24. [PubMed: 21432027]


Hoyer D, Hannon JP, Martin GR. Molecular, pharmacological and functional diversity of 5-HT
receptors. Pharmacol Biochem Behav. 2002; 71:533–554. [PubMed: 11888546]
International Schizophrenia Consortium. Rare chromosomal deletions and duplications increase risk of
schizophrenia. Nature. 2008; 455:237–241. [PubMed: 18668038]
Javitt DC, Lindsley RW. Effects of phencyclidine on prepulse inhibition of acoustic startle response in
the macaque. Psychopharmacology (Berl). 2001; 156:165–168. [PubMed: 11549218]
Jones NC, Reddy M, Anderson P, Salzberg MR, O’Brien TJ, Pinault D. Acute administration of typical
and atypical antipsychotics reduces EEG gamma power, but only the preclinical compound
LY379268 reduces the ketamine-induced rise in gamma power. Int J Neuropsychopharmacol.
2011; 15:657–668. [PubMed: 21733235]
Keeler MH. Similarity of schizophrenia and the psilocybin syndrome as determined by objective
methods. Int J Neuropsychiatry. 1965; 1:630–634. [PubMed: 4380179]
Kellendonk C, Simpson EH, Kandel ER. Modeling cognitive endophenotypes of schizophrenia in
Author Manuscript

mice. Trends Neurosci. 2009; 32:347–358. [PubMed: 19409625]


Khashan AS, Abel KM, McNamee R, Pedersen MG, Webb RT, Baker PN, Kenny LC, Mortensen PB.
Higher risk of offspring schizophrenia following antenatal maternal exposure to severe adverse life
events. Arch Gen Psychiatry. 2008; 65:146–152. [PubMed: 18250252]
Kinon BJ, Zhang L, Millen BA, Osuntokun OO, Williams JE, Kollack-Walker S, Jackson K,
Kryzhanovskaya L, Jarkova N. A multicenter, inpatient, phase 2, double-blind, placebo-controlled
dose-ranging study of LY2140023 monohydrate in patients with DSM-IV schizophrenia. J Clin
Psychopharmacol. 2011; 31:349–355. [PubMed: 21508856]
Klodzinska A, Bijak M, Tokarski K, Pilc A. Group II mGlu receptor agonists inhibit behavioural and
electrophysiological effects of DOI in mice. Pharmacol Biochem Behav. 2002; 73:327–332.
[PubMed: 12117586]
Kozlenkov, A.; Gonzalez-Maeso, J. Animal models and hallucinogenic drugs. In: Jardri, R.; Cachia,
A.; Thomas, R.; Pins, D., editors. The neuroscience of hallucinations. Springer; New York: 2012.
p. 253-277.
Author Manuscript

Krebs-Thomson K, Paulus MP, Geyer MA. Effects of hallucinogens on locomotor and investigatory
activity and patterns: influence of 5-HT2A and 5-HT2C receptors. Neuropsychopharmacology.
1998; 18:339–351. [PubMed: 9536447]
Kristiansen LV, Huerta I, Beneyto M, Meador-Woodruff JH. NMDA receptors and schizophrenia. Curr
Opin Pharmacol. 2007; 7:48–55. [PubMed: 17097347]
Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R, Bremner JD, Heninger GR, Bowers MB Jr.
Charney DS. Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans.
Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry.
1994; 51:199–214. [PubMed: 8122957]
Krystal JH, Abi-Saab W, Perry E, D’Souza DC, Liu N, Gueorguieva R, McDougall L, Hunsberger T,
Belger A, Levine L, Breier A. Preliminary evidence of attenuation of the disruptive effects of the
NMDA glutamate receptor antagonist, ketamine, on working memory by pretreatment with the
group II metabotropic glutamate receptor agonist, LY354740, in healthy human subjects.
Psychopharmacology (Berl). 2005; 179:303–309. [PubMed: 15309376]
Kurita M, et al. HDAC2 regulates atypical antipsychotic responses through the modulation of mGlu2
Author Manuscript

promoter activity. Nat Neurosci. 2012; 15:1245–1254. [PubMed: 22864611]


Lahti AC, Weiler MA, Tamara Michaelidis BA, Parwani A, Tamminga CA. Effects of ketamine in
normal and schizophrenic volunteers. Neuropsychopharmacology. 2001; 25:455–467. [PubMed:
11557159]
Lewis DA, Levitt P. Schizophrenia as a disorder of neurodevelopment. Annu Rev Neurosci. 2002;
25:409–432. [PubMed: 12052915]
Lewis DA, Gonzalez-Burgos G. Pathophysiologically based treatment interventions in schizophrenia.
Nat Med. 2006; 12:1016–1022. [PubMed: 16960576]

Int J Neuropsychopharmacol. Author manuscript; available in PMC 2016 April 19.


Moreno and González-Maeso Page 16

Lewis DA, Curley AA, Glausier JR, Volk DW. Cortical parvalbumin interneurons and cognitive
dysfunction in schizophrenia. Trends Neurosci. 2012; 35:57–67. [PubMed: 22154068]
Author Manuscript

Lieberman JA, Bymaster FP, Meltzer HY, Deutch AY, Duncan GE, Marx CE, Aprille JR, Dwyer DS,
Li XM, Mahadik SP, Duman RS, Porter JH, Modica-Napolitano JS, Newton SS, Csernansky JG.
Antipsychotic drugs: comparison in animal models of efficacy, neurotransmitter regulation, and
neuroprotection. Pharmacol Rev. 2008; 60:358–403. [PubMed: 18922967]
Linden AM, Greene SJ, Bergeron M, Schoepp DD. Anxiolytic activity of the MGLU2/3 receptor
agonist LY354740 on the elevated plus maze is associated with the suppression of stress-induced
c-Fos in the hippocampus and increases in c-Fos induction in several other stress-sensitive brain
regions. Neuropsychopharmacology. 2004; 29:502–513. [PubMed: 14694349]
Linn GS, Javitt DC. Phencyclidine (PCP)-induced deficits of prepulse inhibition in monkeys.
Neuroreport. 2001; 12:117–120. [PubMed: 11201070]
Ludewig K, Geyer MA, Vollenweider FX. Deficits in prepulse inhibition and habituation in never-
medicated, first-episode schizophrenia. Biol Psychiatry. 2003; 54:121–128. [PubMed: 12873801]
Maes M, Song C, Lin A, De Jongh R, Van Gastel A, Kenis G, Bosmans E, De Meester I, Benoy I,
Neels H, Demedts P, Janca A, Scharpe S, Smith RS. The effects of psychological stress on
Author Manuscript

humans: increased production of pro-inflammatory cytokines and a Th1-like response in stress-


induced anxiety. Cytokine. 1998; 10:313–318. [PubMed: 9617578]
Malaspina D, Corcoran C, Kleinhaus KR, Perrin MC, Fennig S, Nahon D, Friedlander Y, Harlap S.
Acute maternal stress in pregnancy and schizophrenia in offspring: a cohort prospective study.
BMC Psychiatry. 2008; 8:71. [PubMed: 18717990]
Mansbach RS, Geyer MA. Effects of phencyclidine and phencyclidine biologs on sensorimotor gating
in the rat. Neuropsychopharmacology. 1989; 2:299–308. [PubMed: 2692589]
Mansbach RS, Geyer MA. Parametric determinants in pre-stimulus modification of acoustic startle:
interaction with ketamine. Psychopharmacology (Berl). 1991; 105:162–168. [PubMed: 1796122]
Markham JA, Koenig JI. Prenatal stress: role in psychotic and depressive diseases.
Psychopharmacology (Berl). 2011; 214:89–106. [PubMed: 20949351]
Marona-Lewicka D, Thisted RA, Nichols DE. Distinct temporal phases in the behavioral
pharmacology of LSD: dopamine D2 receptor-mediated effects in the rat and implications for
psychosis. Psychopharmacology (Berl). 2005; 180:427–435. [PubMed: 15723230]
Author Manuscript

Martin WR, Wikler A, Eades CG, Pescor FT. Tolerance to and physical dependence on morphine in
rats. Psychopharmacologia. 1963; 4:247–260. [PubMed: 14048545]
Matrisciano F, Tueting P, Maccari S, Nicoletti F, Guidotti A. Pharmacological activation of group-II
metabotropic glutamate receptors corrects a schizophrenia-like phenotype induced by prenatal
stress in mice. Neuropsychopharmacology. 2012; 37:929–938. [PubMed: 22089319]
Maurel-Remy S, Bervoets K, Millan MJ. Blockade of phencyclidine-induced hyperlocomotion by
clozapine and MDL 100,907 in rats reflects antagonism of 5-HT2A receptors. Eur J Pharmacol.
1995; 280:R9–11. [PubMed: 7589172]
McFarland K, Price DL, Bonhaus DW. Pimavanserin, a 5-HT2A inverse agonist, reverses psychosis-
like behaviors in a rodent model of Parkinson’s disease. Behav Pharmacol. 2011; 22:681–692.
[PubMed: 21921840]
McGuffin P, Asherson P, Owen M, Farmer A. The strength of the genetic effect. Is there room for an
environmental influence in the aetiology of schizophrenia? Br J Psychiatry. 1994; 164:593–599.
[PubMed: 7921708]
McOmish CE, Lira A, Hanks JB, Gingrich JA. Clozapine-induced locomotor suppression is mediated
Author Manuscript

by 5-HT(2A) receptors in the forebrain. Neuropsychopharmacology. 2012; 37:2747–2755.


[PubMed: 22871913]
Meltzer HY, Alphs L, Green AI, Altamura AC, Anand R, Bertoldi A, Bourgeois M, Chouinard G,
Islam MZ, Kane J, Krishnan R, Lindenmayer JP, Potkin S. Clozapine treatment for suicidality in
schizophrenia: international suicide prevention trial (InterSePT). Arch Gen Psychiatry. 2003a;
60:82–91. [PubMed: 12511175]
Meltzer HY, Li Z, Kaneda Y, Ichikawa J. Serotonin receptors: their key role in drugs to treat
schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry. 2003b; 27:1159–1172. [PubMed:
14642974]

Int J Neuropsychopharmacol. Author manuscript; available in PMC 2016 April 19.


Moreno and González-Maeso Page 17

Meltzer HY, Bonaccorso S, Bobo WV, Chen Y, Jayathilake K. A 12-month randomized, open-label
study of the metabolic effects of olanzapine and risperidone in psychoticpatients: influence of
Author Manuscript

valproic acid augmentation. J Clin Psychiatry. 2011; 72:1602–1610. [PubMed: 21813074]


Menninger KA. Psychoses associated with influenza. J Am Med Assoc. 1919; 72:235–241.
Mestre TA, Zurowski M, Fox SH. 5-Hydroxytryptamine 2A receptor antagonists as potential treatment
for psychiatric disorders. Expert Opin Investig Drugs. 2013; 22:411–421.
Minzenberg MJ, Carter CS. Developing treatments for impaired cognition in schizophrenia. Trends
Cogn Sci. 2012; 16:35–42. [PubMed: 22178120]
Mittal VA, Ellman LM, Cannon TD. Gene-environment interaction and covariation in schizophrenia:
the role of obstetric complications. Schizophr Bull. 2008; 34:1083–1094. [PubMed: 18635675]
Miyamoto S, Duncan GE, Marx CE, Lieberman JA. Treatments for schizophrenia: a critical review of
pharmacology and mechanisms of action of antipsychotic drugs. Mol Psychiatry. 2005; 10:79–
104. [PubMed: 15289815]
Moghaddam B, Adams BW. Reversal of phencyclidine effects by a group II metabotropic glutamate
receptor agonist in rats. Science. 1998; 281:1349–1352. [PubMed: 9721099]
Moreno JL, Holloway T, Albizu L, Sealfon SC, Gonzalez-Maeso J. Metabotropic glutamate mGlu2
Author Manuscript

receptor is necessary for the pharmacological and behavioral effects induced by hallucinogenic 5-
HT2A receptor agonists. Neurosci Lett. 2011a; 493:76–79. [PubMed: 21276828]
Moreno JL, Kurita M, Holloway T, Lopez J, Cadagan R, Martinez-Sobrido L, Garcia-Sastre A,
Gonzalez-Maeso J. Maternal influenza viral infection causes schizophrenia-like alterations of 5-
HT2A and mGlu2 receptors in the adult offspring. J Neurosci. 2011b; 31:1863–1872. [PubMed:
21289196]
Moreno JL, Muguruza C, Umali A, Mortillo S, Holloway T, Pilar-Cuellar F, Mocci G, Seto J, Callado
LF, Neve RL, Milligan G, Sealfon SC, Lopez-Gimenez JF, Meana JJ, Benson DL, Gonzalez-
Maeso J. Identification of three residues essential for 5-HT2A-mGlu2 receptor heteromerization
and its psychoactive behavioral function. J Biol Chem. 2012; 287:44301–44319. [PubMed:
23129762]
Moreno JL, Holloway T, Rayannavar V, Sealfon SC, Gonzalez-Maeso J. Chronic treatment with
LY341495 decreases 5-HT(2A) receptor binding and hallucinogenic effects of LSD in mice.
Neurosci Lett. 2013a; 536:69–73. [PubMed: 23333599]
Author Manuscript

Moreno JL, Holloway T, Umali A, Rayannavar V, Sealfon SC, Gonzalez-Maeso J. Persistent effects of
chronic clozapine on the cellular and behavioral responses to LSD in mice. Psychopharmacology
(Berl). 2013b; 225:217–226. [PubMed: 22842765]
Morris BJ, Cochran SM, Pratt JA. PCP: from pharmacology to modelling schizophrenia. Curr Opin
Pharmacol. 2005; 5:101–106. [PubMed: 15661633]
Muguruza C, Moreno JL, Umali A, Callado LF, Meana JJ, Gonzalez-Maeso J. Dysregulated 5-HT2A
receptor binding in postmortem frontal cortex of schizophrenic subjects. Eur
Neuropsychopharmacol. 2012 Advance online publication. doi: 10.1016/j.euroneuro.
2012.10.006.
Nawa H, Takei N. Recent progress in animal modeling of immune inflammatory processes in
schizophrenia: implication of specific cytokines. Neurosci Res. 2006; 56:2–13. [PubMed:
16837094]
Nestler EJ, Hyman SE. Animal models of neuropsychiatric disorders. Nat Neurosci. 2010; 13:1161–
1169. [PubMed: 20877280]
Newcomer JW, Farber NB, Jevtovic-Todorovic V, Selke G, Melson AK, Hershey T, Craft S, Olney JW.
Author Manuscript

Ketamine-induced NMDA receptor hypofunction as a model of memory impairment and


psychosis. Neuropsychopharmacology. 1999; 20:106–118. [PubMed: 9885791]
Nichols DE. Hallucinogens. Pharmacol Ther. 2004; 101:131–181. [PubMed: 14761703]
Nicodemus KK, Marenco S, Batten AJ, Vakkalanka R, Egan MF, Straub RE, Weinberger DR. Serious
obstetric complications interact with hypoxia-regulated/vascular-expression genes to influence
schizophrenia risk. Mol Psychiatry. 2008; 13:873–877. [PubMed: 18195713]
Ninan I, Kulkarni SK. 5-HT2A receptor antagonists block MK-801-induced stereotypy and
hyperlocomotion. Eur J Pharmacol. 1998; 358:111–116. [PubMed: 9808259]

Int J Neuropsychopharmacol. Author manuscript; available in PMC 2016 April 19.


Moreno and González-Maeso Page 18

O’Neill MF, Hicks CA, Shaw G, Parameswaran T, Cardwell GP, O’Neill MJ. Effects of 5-
hydroxytryptamine2 receptor antagonism on the behavioral activation and immediate early gene
Author Manuscript

expression induced by dizocilpine. J Pharmacol Exp Ther. 1998; 287:839–846. [PubMed:


9864262]
O’Tuathaigh CM, Kirby BP, Moran PM, Waddington JL. Mutant mouse models: genotype-phenotype
relationships to negative symptoms in schizophrenia. Schizophr Bull. 2009; 36:271–288.
[PubMed: 19934211]
Ouagazzal A, Grottick AJ, Moreau J, Higgins GA. Effect of LSD on prepulse inhibition and
spontaneous behavior in the rat. A pharmacological analysis and comparison between two rat
strains. Neuropsychopharmacology. 2001; 25:565–575. [PubMed: 11557170]
Patil ST, Zhang L, Martenyi F, Lowe SL, Jackson KA, Andreev BV, Avedisova AS, Bardenstein LM,
Gurovich IY, Morozova MA, Mosolov SN, Neznanov NG, Reznik AM, Smulevich AB, Tochilov
VA, Johnson BG, Monn JA, Schoepp DD. Activation of mGlu2/3 receptors as a new approach to
treat schizophrenia: a randomized Phase 2 clinical trial. Nat Med. 2007; 13:1102–1107.
[PubMed: 17767166]
Patterson PH. Maternal infection and immune involvement in autism. Trends Mol Med. 2011; 17:389–
Author Manuscript

394. [PubMed: 21482187]


Pearlson GD. Psychiatric and medical syndromes associated with phencyclidine (PCP) abuse. Johns
Hopkins Med J. 1981; 148:25–33. [PubMed: 6109037]
Pompili M, Amador XF, Girardi P, Harkavy-Friedman J, Harrow M, Kaplan K, Krausz M, Lester D,
Meltzer HY, Modestin J, Montross LP, Mortensen PB, Munk-Jorgensen P, Nielsen J, Nordentoft
M, Saarinen PI, Zisook S, Wilson ST, Tatarelli R. Suicide risk in schizophrenia: learning from the
past to change the future. Ann Gen Psychiatry. 2007; 6:10. [PubMed: 17367524]
Powell CM, Miyakawa T. Schizophrenia-relevant behavioral testing in rodent models: a uniquely
human disorder? Biol Psychiatry. 2006; 59:1198–1207. [PubMed: 16797265]
Powell SB, Geyer MA. Overview of animal models of schizophrenia. Curr Protoc Neurosci. 2007
Chapter 9:Unit 9.24.
Powell SB, Palomo J, Carasso BS, Bakshi VP, Geyer MA. Yohimbine disrupts prepulse inhibition in
rats via action at5-HT1A receptors, not alpha2-adrenoceptors. Psychopharmacology (Berl). 2005;
180:491–500. [PubMed: 15719216]
Pratt JA, Winchester C, Egerton A, Cochran SM, Morris BJ. Modelling prefrontal cortex deficits in
Author Manuscript

schizophrenia: implications for treatment. Br J Pharmacol. 2008; 153(Suppl. 1):S465–S470.


[PubMed: 18311160]
Purcell SM, Wray NR, Stone JL, Visscher PM, O’Donovan MC, Sullivan PF, Sklar P. Common
polygenic variation contributes to risk of schizophrenia and bipolar disorder. Nature. 2009;
460:748–752. [PubMed: 19571811]
Quednow BB, Kometer M, Geyer MA, Vollenweider FX. Psilocybin-induced deficits in automatic and
controlled inhibition are attenuated by ketanserin in healthy human volunteers.
Neuropsychopharmacology. 2011; 37:630–640. [PubMed: 21956447]
Rainey JM Jr. Crowder MK. Prolonged psychosis attributed to phencyclidine: report of three cases.
Am J Psychiatry. 1975; 132:1076–1078. [PubMed: 1166881]
Ross CA, Margolis RL, Reading SA, Pletnikov M, Coyle JT. Neurobiology of schizophrenia. Neuron.
2006; 52:139–153. [PubMed: 17015232]
Saha S, Chant D, McGrath J. A systematic review of mortality in schizophrenia: is the differential
mortality gap worsening over time? Arch Gen Psychiatry. 2007; 64:1123–1131. [PubMed:
Author Manuscript

17909124]
Sawa A, Snyder SH. Schizophrenia: diverse approaches to a complex disease. Science. 2002; 296:692–
695. [PubMed: 11976442]
Schmidt A, Diaconescu AO, Kometer M, Friston KJ, Stephan KE, Vollenweider FX. Modeling
ketamine effects on synaptic plasticity during the mismatch negativity. Cereb Cortex. 2012
Advance online publication. doi:10.1093/cercor/bhs238.
Schmidt A, Kometer M, Bachmann R, Seifritz E, Vollenweider F. The NMDA antagonist ketamine and
the 5-HT agonist psilocybin produce dissociable effects on structural encoding of emotional face
expressions. Psychopharmacology (Berl). 2013; 225:227–239. [PubMed: 22836372]

Int J Neuropsychopharmacol. Author manuscript; available in PMC 2016 April 19.


Moreno and González-Maeso Page 19

Sipes TE, Geyer MA. DOI disrupts prepulse inhibition of startle in rats via 5-HT2A receptors in the
ventral pallidum. Brain Res. 1997; 761:97–104. [PubMed: 9247071]
Author Manuscript

Sorensen HJ, Mortensen EL, Reinisch JM, Mednick SA. Association between prenatal exposure to
bacterial infection and risk of schizophrenia. Schizophr Bull. 2009; 35:631–637. [PubMed:
18832344]
Stefansson H, et al. Large recurrent microdeletions associated with schizophrenia. Nature. 2008;
455:232–236. [PubMed: 18668039]
Stefansson H, et al. Common variants conferring risk of schizophrenia. Nature. 2009; 460:744–747.
[PubMed: 19571808]
Steptoe A, Willemsen G, Owen N, Flower L, Mohamed-Ali V. Acute mental stress elicits delayed
increases in circulating inflammatory cytokine levels. Clin Sci (Lond). 2001; 101:185–192.
[PubMed: 11473494]
Susser E, St Clair D, He L. Latent effects of prenatal malnutrition on adult health: the example of
schizophrenia. Ann N Y Acad Sci. 2008; 1136:185–192. [PubMed: 18579882]
Swerdlow NR, Stephany N, Wasserman LC, Talledo J, Shoemaker J, Auerbach PP. Amphetamine
effects on prepulse inhibition across-species: replication and parametric extension.
Author Manuscript

Neuropsychopharmacology. 2003; 28:640–650. [PubMed: 12655308]


Tamminga CA, Holcomb HH. Phenotype of schizophrenia: a review and formulation. Mol Psychiatry.
2005; 10:27–39. [PubMed: 15340352]
Tsuang MT, Stone WS, Faraone SV. Genes, environment and schizophrenia. Br J Psychiatry Suppl.
2001; 40:s18–s24. [PubMed: 11315219]
Umbricht D, Schmid L, Koller R, Vollenweider FX, Hell D, Javitt DC. Ketamine-induced deficits in
auditory and visual context-dependent processing in healthy volunteers: implications for models
of cognitive deficits in schizophrenia. Arch Gen Psychiatry. 2000; 57:1139–1147. [PubMed:
11115327]
Umbricht D, Koller R, Vollenweider FX, Schmid L. Mismatch negativity predicts psychotic
experiences induced by NMDA receptor antagonist in healthy volunteers. Biol Psychiatry. 2002;
51:400–406. [PubMed: 11904134]
Uslaner JM, Smith SM, Huszar SL, Pachmerhiwala R, Hinchliffe RM, Vardigan JD, Hutson PH.
Combined administration of an mGlu2/3 receptor agonist and a 5-HT 2A receptor antagonist
Author Manuscript

markedly attenuate the psychomotor-activating and neurochemical effects of psychostimulants.


Psychopharmacology. 2009; 206:641–651. [PubMed: 19707745]
Vacic V, et al. Duplications of the neuropeptide receptor gene VIPR2 confer significant risk for
schizophrenia. Nature. 2011; 471:499–503. [PubMed: 21346763]
Vanbesien-Mailliot CC, Wolowczuk I, Mairesse J, Viltart O, Delacre M, Khalife J, Chartier-Harlin
MC, Maccari S. Prenatal stress has pro-inflammatory consequences on the immune system in
adult rats. Psychoneuroendocrinology. 2007; 32:114–124. [PubMed: 17240075]
van den Buuse M. Modeling the positive symptoms of schizophrenia in genetically modified mice:
pharmacology and methodology aspects. Schizophr Bull. 2010; 36:246–270. [PubMed:
19900963]
Van Oekelen D, Megens A, Meert T, Luyten WH, Leysen JE. Functional study of rat 5-HT2A
receptors using antisense oligonucleotides. J Neurochem. 2003; 85:1087–1100. [PubMed:
12753068]
van Os J, Kapur S. Schizophrenia. Lancet. 2009; 374:635–645. [PubMed: 19700006]
van Os J, Selten JP. Prenatal exposure to maternal stress and subsequent schizophrenia. The May 1940
Author Manuscript

invasion of The Netherlands. Br J Psychiatry. 1998; 172:324–326. [PubMed: 9715334]


Vollenweider FX, Leenders KL, Oye I, Hell D, Angst J. Differential psychopathology and patterns of
cerebral glucose utilisation produced by (S)- and (R)-ketamine in healthy volunteers using
positron emission tomography (PET). Eur Neuropsychopharmacol. 1997a; 7:25–38. [PubMed:
9088882]
Vollenweider FX, Leenders KL, Scharfetter C, Antonini a, Maguire P, Missimer J, Angst J. Metabolic
hyperfrontality and psychopathology in the ketamine model of psychosis using positron emission
tomography (PET) and [18F]fluorodeoxyglucose (FDG). Eur Neuropsychopharmacol. 1997b;
7:9–24. [PubMed: 9088881]

Int J Neuropsychopharmacol. Author manuscript; available in PMC 2016 April 19.


Moreno and González-Maeso Page 20

Vollenweider FX, Vollenweider-Scherpenhuyzen MF, Babler A, Vogel H, Hell D. Psilocybin induces


schizophrenia-like psychosis in humans via a serotonin-2 agonist action. Neuroreport. 1998;
Author Manuscript

9:3897–3902. [PubMed: 9875725]


Vollenweider FX, Vontobel P, Oye I, Hell D, Leenders KL. Effects of (S)-ketamine on striatal
dopamine: a [11C] raclopride PET study of a model psychosis in humans. J Psychiatr Res. 2000;
34:35–43. [PubMed: 10696831]
Walsh T, et al. Rare structural variants disrupt multiple genes in neurodevelopmental pathways in
schizophrenia. Science. 2008; 320:539–543. [PubMed: 18369103]
Williams AA, Ingram WM, Levine S, Resnik J, Kamel CM, Lish JR, Elizalde DI, Janowski SA,
Shoker J, Kozlenkov A, Gonzalez-Maeso J, Gallitano AL. Reduced levels of serotonin 2A
receptors underlie resistance of Egr3-deficient mice to locomotor suppression by clozapine.
Neuropsychopharmacology. 2012; 37:2285–2298. [PubMed: 22692564]
Willins DL, Meltzer HY. Direct injection of 5-HT2A receptor agonists into the medial prefrontal
cortex produces a head-twitch response in rats. J Pharmacol Exp Ther. 1997; 282:699–706.
[PubMed: 9262333]
Winter JC, Eckler JR, Rabin RA. Serotonergic/glutamatergic interactions: the effects of mGlu2/3
Author Manuscript

receptor ligands in rats trained with LSD and PCP as discriminative stimuli.
Psychopharmacology (Berl). 2004; 172:233–240. [PubMed: 14598016]
Woolley ML, Pemberton DJ, Bate S, Corti C, Jones DN. The mGlu2 but not the mGlu3 receptor
mediates the actions of the mGluR2/3 agonist, LY379268, in mouse models predictive of
antipsychotic activity. Psychopharmacology (Berl). 2008; 196:431–440. [PubMed: 18057917]
Yadav PN, Abbas AI, Farrell MS, Setola V, Sciaky N, Huang XP, Kroeze WK, Crawford LK, Piel DA,
Keiser MJ, Irwin JJ, Shoichet BK, Deneris ES, Gingrich J, Beck SG, Roth BL. The presynaptic
component of the serotonergic system is required for clozapine’s efficacy.
Neuropsychopharmacology. 2011a; 36:638–651. [PubMed: 21048700]
Yadav PN, Kroeze WK, Farrell MS, Roth BL. Antagonist functional selectivity: 5-HT2A serotonin
receptor antagonists differentially regulate 5-HT2A receptor protein level in vivo. J Pharmacol
Exp Ther. 2011b; 339:99–105. [PubMed: 21737536]
Yolken RH, Torrey EF. Are some cases of psychosis caused by microbial agents? A review of the
evidence. Mol Psychiatry. 2008; 13:470–479. [PubMed: 18268502]
Young BG. A phenomenological comparison of LSD and schizophrenic states. Br J Psychiatry. 1974;
Author Manuscript

124:64–74. [PubMed: 4822416]


Yudofsky SC. Contracting schizophrenia: lessons from the influenza epidemic of 1918–1919. JAMA.
2009; 301:324–326. [PubMed: 19155461]
Author Manuscript

Int J Neuropsychopharmacol. Author manuscript; available in PMC 2016 April 19.

You might also like