Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Mar 12.
Published in final edited form as: Curr Top Behav Neurosci. 2010;4:435–481. doi: 10.1007/7854_2010_57

Models of Neurodevelopmental Abnormalities in Schizophrenia

Susan B Powell 1
PMCID: PMC3595002  NIHMSID: NIHMS443787  PMID: 21312409

Abstract

The neurodevelopmental hypothesis of schizophrenia asserts that the underlying pathology of schizophrenia has its roots in brain development and that these brain abnormalities do not manifest themselves until adolescence or early adulthood. Animal models based on developmental manipulations have provided insight into the vulnerability of the developing fetus and the importance of the early environment for normal maturation. These models have provided a wide range of validated approaches to answer questions regarding environmental influences on both neural and behavioral development. In an effort to better understand the developmental hypothesis of schizophrenia, animal models have been developed, which seek to model the etiology and/or the pathophysiology of schizophrenia or specific behaviors associated with the disease. Developmental models specific to schizophrenia have focused on epidemiological risk factors (e.g., prenatal viral insult, birth complications) or more heuristic models aimed at understanding the developmental neuropathology of the disease (e.g., ventral hippocampal lesions). The combined approach of behavioral and neuroanatomical evaluation of these models strengthens their utility in improving our understanding of the pathophysiology of schizophrenia and developing new treatment strategies.

Keywords: Development, Immune, Social isolation, Stress, Neonatal ventral hippocampal lesion, Protein deprivation, Prenatal, Neonatal, Postnatal, Animal model, Schizophrenia, Toxin, Obstetric complications, Behavior

1 Neurodevelopmental Models of Schizophrenia

1.1 Developmental Theory of Schizophrenia

Over the past two decades, development of the central nervous system has become critical in understanding the neurobiology of schizophrenia (Fatemi and Folsom 2009; Lewis and Levitt 2002; Murray and Lewis 1988; Weinberger 1987). The neurodevelopmental hypothesis of schizophrenia asserts that the underlying pathology of schizophrenia has its roots in brain development and that these brain abnormalities do not manifest themselves until adolescence or early adulthood (Fatemi and Folsom 2009; Rapoport et al. 2005). In addition to the course of illness, support for a neurodevelopmental etiology comes from neuroanatomical and cytoarchitectural abnormalities in the brains of patients with schizophrenia. For example, ventricular enlargement and decreased cortical, hippocampal, and amygdalar volumes are present without any evidence of gliosis (i.e., trauma or neurodegeneration, Arnold et al. 1997; Fatemi and Folsom 2009; Weinberger 1987). Additionally, misplaced and clustered neurons, particularly in the entorhinal cortex, indicate problems of neuronal migration and suggest an early developmental anomaly (Arnold et al. 1991; Falkai et al. 2000; Jakob and Beckmann 1986). Pyramidal neurons in the hippocampus and neocortex have smaller cell bodies and fewer dendritic spines and dendritic arborizations (reviewed in Harrison and Weinberger 2005). Additionally, decreased presynaptic proteins such as synapto-physin, SNAP-25, and complexin II have been observed in schizophrenia brains as well as decreased density of interneurons (e.g., parvalbumin-immunoreactive cells) (Harrison and Weinberger 2005). There are also reports of decreases in cell numbers in the thalamus and a decreased number of oligodendrocytes. Neuroimaging data and postmortem studies have shown that N-acetylaspartate (NAA), a marker of neuronal integrity, is decreased in first episode and in never-medicated patients (Bertolino and Weinberger 1999; Nudmamud et al. 2003). On the basis of these neuropathological changes, investigators have conceptualized schizophrenia as a disease of functional “dysconnectivity” (Friston and Frith 1995; McGlashan and Hoffman 2000; Weinberger et al. 1992) or a “disorder of the synapse” affecting the machinery of the synapse (Frankle et al. 2003; reviewed in Harrison and Weinberger 2005; Mirnics et al. 2001). Recent evidence from MRI studies of reduced white matter supports the disconnection model of schizophrenia (Fatemi and Folsom 2009).

Epidemiological studies support the notion that environmental factors contribute to the incidence of schizophrenia (Cannon et al. 2003; Rapoport et al. 2005). For example, season of birth is a risk factor, with late winter/early spring births associated with an increased risk of schizophrenia (Boyd et al. 1986; Machon et al. 1983; Mino and Oshima 2006; Torrey et al. 1997). Recent studies showed that social factors such as urbanicity, immigrant status, and social isolation are associated with an increased risk for schizophrenia (Cannon et al. 2008; Dean et al. 2003; Marcelis et al. 1998). Hence, the developmental hypothesis of schizophrenia has led to the examination of environmental and epigenetic factors associated with schizophrenia, asserting that an early environmental insult, such as a viral exposure to the developing fetus (Brown and Susser 2002; Mednick et al. 1988; O’Callaghan et al. 1991; Takei et al. 1996) or obstetric complications (Owen and Lewis 1988), causes dysfunction in neural systems that normally reach maturity in late adolescence and early adulthood. Thus, the symptoms of schizophrenia would not express themselves until the point in development in which these brain areas (e.g., dorsal prefrontal cortex) mature (Weinberger 1987).

1.2 Animal Models of Developmental Hypothesis

Several animal models are being used to understand neurobiological processes relevant to the developmental hypothesis of schizophrenia (Fatemi and Folsom 2009; Lipska and Weinberger 2000; Meyer and Feldon 2009a, b; Powell and Geyer 2002). Although recreating a uniquely human condition such as schizophrenia is not feasible in animals, animal models have been useful in aiding our understanding of the pathophysiology of the disease (Geyer and Markou 2002; Powell and Geyer 2007; Swerdlow et al. 1994) (Young et al. 2010). These animal models are evaluated based on their face, construct, and predictive validity, and involve both the manipulations (e.g., pharmacological, developmental insult) and measures (e.g., prepulse inhibition, cognitive flexibility). For a more detailed description of the differences in various forms of validity, see Young et al. this text, Geyer and Markou (2002), Swerdlow et al. (1994). When evaluating the relevance of the model to schizophrenia, one should consider both the manipulations and the measures.

Developmental models specific to schizophrenia have focused on the intrauterine environment [e.g., viral insult, exposure to neurotoxins, prenatal maternal stress; (Fatemi et al. 2005; Meyer et al. 2009)], birth complications [e.g., cesarean section, hypoxia; (Boksa and El-Khodor 2003; Vaillancourt and Boksa 2000; Wakuda et al. 2008)], perinatal insult [e.g., ventral hippocampal lesions (Tseng et al. 2009)], prenatal stress (Koenig et al. 2002), and postnatal maternal and/or social deprivation (Ellenbroek et al. 1998; Fone and Porkess 2008; Powell and Geyer 2002). Additionally, genetic models, particularly those targeting developmental genes or those that display age-dependent emergence of a phenotype, can also address neurodevelopmental aspects of schizophrenia (Powell et al. 2009). The combined approach of behavioral and neuroanatomical evaluation of these models strengthens their utility in improving our understanding of the pathophysiology of schizophrenia and developing new treatment strategies (Meyer and Feldon 2009a) including prophylaxis (Meyer et al. 2008d; Powell et al. 2003).

There are several specific experimental factors that need to be considered when conducting developmental studies such as comparisons of the time course of CNS maturation across species, timing of the environmental manipulation, litter effects, and cross-fostering. For a more detailed review of these experimental considerations, see Meyer and Feldon (2009a). Rats and mice differ greatly from humans in the timing of brain development, with brain development in rodents occurring at a much faster pace than that in humans. Considering the proportion of time relative to the lifespan of the organism and the timing of specific neuronal processes, late gestation in humans most likely corresponds to the early postnatal period in rats and mice (Clancy et al. 2001, 2007). Comparisons in brain development between species can be estimated using an algorithm, originally described by (Finlay and Darlington 1995), which is now accessible through a web site (http://translatingtime.net/) (Clancy et al. 2007). Thus, the nature of the schizophrenia-relevant risk factor or neuropathology being modeled and the timing of insult are important factors to consider when evaluating neurodevelopmental models of schizophrenia.

Other important experimental considerations to take into account are in utero environment and maternal behavior. In prenatal challenge models, cross-fostering involves transferring pups from one dam to another lactating surrogate dam to account for any effects the challenge had on mother–pup interactions. For example, in order to confirm significant effects of your manipulation (e.g., prenatal immune activation) it is important to rule out effects of maternal behavior on the observed experimental results. An additional experimental consideration in developmental experiments is litter effects. In multiparous species such as rats and mice, there are often anywhere from 6 to 12 pups born at the same time. This fecundity, while one of the main reasons that rodents are the preferred laboratory species, presents problems to experimental design and statistical analyses, particularly in developmental studies (reviewed in Zorrilla 1997). Owing to shared genes, intrauterine environment, and common postnatal environment, littermates are more similar to each other than nonlittermates and are thus not independent observations. This interdependence complicates statistical analyses and, when each littermate is treated as an independent sample in the ANOVA, inflates the sample size and increases the likelihood of observing a false positive or a false negative (Zorrilla 1997). There are several ways to handle litter effects in the experimental design and statistical analysis, which are discussed in the final section of this chapter and in Zorrilla (1997).

2 Behavioral Measures

Several behavioral measures with certain degrees of validity have been used to assess neurodevelopmental manipulations of relevance to schizophrenia. In this chapter, we will focus primarily on the developmental manipulations themselves and report on the subsequent behavioral and neuronal abnormalities produced by the manipulation. For a more complete review of the “measures” or behavioral tasks used in animal models of schizophrenia, the reader is referred to Jones et al. (2008), Powell and Geyer (2007), Young et al. (2009), Young et al. this text. Briefly, developmental models have been evaluated across several behaviors of relevance to schizophrenia. Generally, these measures fall into four categories: locomotor activity (e.g., spontaneous and drug-induced), gating (e.g., prepulse inhibition of startle, auditory gating), cognitive (e.g., learning and memory, behavioral flexibility), and social (e.g., social interaction, social recognition).

2.1 Spontaneous and Drug-Induced Locomotor Activity

Measures of locomotion and stereotypy in animals have been useful in the identification of drugs that treat the positive symptoms of schizophrenia (e.g., dopamine D2 receptor antagonists). Typically, these experiments involve administering a psychostimulant such as amphetamine to a rat or mouse and observing both the quantity and quality of motor activity produced (reviewed in Segal and Geyer 1985; Segal et al. 1981; Swerdlow et al. 1986). In the absence of a pharmacological manipulation, as is the case with developmental models, spontaneous locomotor activity is often used to evaluate exploratory behavior and unconditioned anxiety produced by a developmental insult. Many of the developmental models reviewed here have also been evaluated for drug-induced locomotor activity (e.g., response to amphetamine, phencyclidine) to probe the functional integrity of the dopamine and glutamate systems, respectively. The measurement of amphetamine-induced locomotor activity is based on the more general dopamine hyperactivity hypothesis of schizophrenia and more specifically, the finding that patients with schizophrenia show exaggerated dopamine release and an exacerbation of symptoms in response to amphetamine (Laruelle et al. 1996).

2.2 Gating Deficits

Deficient gating of sensory input or intrusive thoughts in schizophrenia patients has been recognized for a number of years (Kietzman et al. 1985). In experimental animal studies, gating deficits are evaluated using three primary measures: prepulse inhibition (PPI) of startle, auditory gating, and latent inhibition (LI). Perhaps, the most common behavioral measure assessed in animal models of schizophrenia is PPI of the startle response, an operational measure of sensorimotor gating [reviewed in (Swerdlow and Geyer 1998; Swerdlow et al. 2008)]. PPI is disrupted in schizophrenia patients (Braff et al. 2001) (See Braff 2010) and in pharmacological, developmental, and genetic animal models of schizophrenia (Geyer et al. 2001; Powell and Geyer 2002; Powell et al. 2009). PPI is reliable, can be tested repeatedly in the same animal and has demonstrated face, construct, and predictive validity in animal models of schizophrenia (Geyer and Moghaddam 2002; Swerdlow et al. 1994, 2008). The ability to test the same behavior in the same animals repeatedly is a particularly attractive feature for developmental models that involve the assessment of behaviors both pre- and postpuberty. Together with amphetamine stimulated locomotor activity, sensorimotor gating, as measured by PPI, is often the “gold standard” behavioral endpoint in neurodevelopmental models of schizophrenia research.

The auditory “sensory gating” paradigm in animal studies is based on a similar paired-stimulus paradigm in humans in which the P50 event-related potential (ERP) elicited by the second of two audible clicks is normally reduced relative to the ERP elicited by the first click (Freedman et al. 1999). Because schizophrenia patients do now show the normal reduction in ERP to the second click, a rodent version based on the N40 ERP generated from the hippocampus has been evaluated in animal models of schizophrenia (Freedman et al. 1999; Stevens et al. 1997). LI is conceptually related to the gating theories of schizophrenia disorders and refers to the observation that repeated exposures to a sensory stimulus (i.e., habituation) retards the rate at which a subject will subsequently acquire a stimulus–response association based on this stimulus (Weiner and Arad 2009; Weiner et al. 1988). Meyer et al. (2005) hypothesized that decreases in LI may reflect increased distraction by irrelevant stimuli.

2.3 Attention

Attentional problems in schizophrenia are among the core features of the disease (Addington et al. 1997). In laboratory tests, schizophrenia patients show deficits in the continuous performance task (CPT), which measures sustained attention (Nestor and O’Donnell 1998; Orzack and Kornetsky 1966). Rodent models of attention include the 5-choice serial reaction time task (5-CSRTT) developed by Robbins and colleagues (Chudasama and Robbins 2004; Robbins 2002). Other rodent tasks of attention include the sustained attention task pioneered by Sarter and colleagues (Sarter et al. 2001). For a more complete review of attentional tasks in animal models of schizophrenia, see Young et al. (2009).

2.4 Cognitive Deficits

Because cognitive deficits in schizophrenia are part of the core features of the illness, are associated with poor quality of life, and are relatively resistant to current treatments, there is a renewed focus on defining and treating cognitive deficits (Green et al. 2004; Nuechterlein et al. 2004). The cognitive deficits in schizophrenia include impairments in working memory as well as problem solving, social cognition, and learning and memory (Cannon et al. 2005; Hagan and Jones 2005; Nuechterlein et al. 2004). Animal models mapping onto the specific cognitive domains deficient in schizophrenia are extensively reviewed elsewhere (Young et al. 2009). Briefly, rodent models have focused primarily on assessments of learning and memory (e.g., novel object recognition, Morris water maze; fear conditioning), working memory (e.g., delayed alternation in T-maze), and cognitive flexibility (e.g., set shifting, reversal learning; (Floresco et al. 2009). These tasks are outlined in the previous section (Young et al. this text) and in a recent review paper (Young et al. 2009). Important to developmental models of schizophrenia is the observation that cognitive deficits (e.g., processing speed, working memory, executive function, verbal memory) often predate the onset of psychotic symptoms (Eastvold et al. 2007). This early emergence of cognitive deficits in the prodromal phase should be considered in relation to the postpubertal emergence criteria adopted by many neurodevelopmental animal models. Thus, the emergence of early deficits in cognitive function in an animal model could strengthen its usefulness, particularly in relation to early intervention studies aimed at modeling prodromal treatments.

2.5 Social Interaction

Social withdrawal is included among the negative symptoms of schizophrenia and is often one of the earliest symptoms to occur (Johnstone et al. 2005; McClellan et al. 2003; Miller et al. 2002). Animal models of social impairments fall into three primary categories: social interaction allowing contact between the animal, social approach without contact, and social recognition/social novelty. Social interaction models involve exposing a rat to a nonaggressive conspecific and scoring the amount and type of social interaction [e.g., rough and tumble play, allogrooming; (Sams-Dodd 1996, 1998)]. Rodent social interaction tests such as these have shown their usefulness as a screen for putative antipsychotic medications (e.g., Bruins Slot et al. 2005). A simple test of social approach and novelty was established recently by Crawley, Moy, and colleagues. In this paradigm, test mice are placed in a three-chambered arena, in one chamber, a “stranger” mouse is placed under a wire cup, and in the opposite chamber there is an empty wire container (Crawley 2007; Moy et al. 2004). Exploratory behavior of the test mouse is quantified for 10 min, with social approach measured by comparing the number of contacts and time spent at the container with the “stranger” mouse compared to the empty container. After 10 min, social novelty is tested by putting a new mouse into the previously empty container, and comparing the exploration of the stranger mouse explored in the approach test and the new mouse. This measure of “social novelty” is very similar to other social recognition tests, which involve assessing the time spent investigating a novel, unfamiliar conspecific in the presence of a familiar conspecific (Engelmann et al. 1995; Ferguson et al. 2002; Thor and Holloway 1981; Winslow and Camacho 1995).

3 Epidemiologic-Based Developmental Manipulations

Historically, the manipulations used to produce animal models involved assessment of drug-induced changes in behavior, particularly in response to psychotomimetic drugs (e.g., amphetamine-induced hyperactivity, phencyclidine-induced PPI disruptions). While pharmacological models have proven useful in the evaluation of antipsychotics, developmental manipulations offer the unique ability to probe etiological factors associated with schizophrenia and provide models for the assessment of novel therapeutics (Meyer and Feldon 2009a). For example, assessment of drugs in amphetamine-induced hyperactivity model will tend to screen for compounds predominantly on dopamine antagonist properties similar to currently available antipsychotics. Neurodevelopmental models can be either epidemiologic and focus on the specific risk factors in the human populations (e.g., prenatal infection) (for review, see Meyer and Feldon 2009a) or be heuristic models of developmental neuropathology observed in schizophrenia (e.g., neonatal ventral hippocampal lesion) (for review, see Tseng et al. 2009). Hence, for the purposes of this discussion, the relevant neurodevelopmental animal models of schizophrenia are divided into two categories: epidemiologic and heuristic models (Table 1). Of course, there is some overlap between the two categories and it can be debated how well each model represents the epidemiological risk factor and/or neuropathology it attempts to address. Nevertheless, this distinction is used to guide the discussion of the developmental models reviewed in this chapter.

Table 1.

Overview of neurodevelopmental models

Risk factor/pathology Rodent model References
Epidemiological models
Maternal infection Prenatal viral infection Shi et al. (2003), Fatemi et al. (1998, 2002a, b, 2004)
Prenatal PolyI:C Shi et al. (2003), Meyer et al. (2009)a, Meyer and Feldon (2009b)a
Prenatal LPS Borrell et al. (2002), Romero et al. (2007, 2008), Fortier et al. (2004)
Prenatal cytokine Smith et al. (2007)
Neonatal infection Neonatal viral infection Asp et al. (2009), Rothschild et al. (1999)
Neonatal cytokine exposure Tohmi et al. (2004), Tsuda et al. (2006), Watanabe et al. (2004)
Neonatal PolyI:C Ibi et al. (2009)
Neonatal LPS Jenkins et al. (2009)
Prenatal stress/maternal deprivation Prenatal restraint stress Gue et al. (2004), Kapoor et al. (2009), Lemaire et al. (2000), Son et al. (2006), Szuran et al. (2000), Wu et al. (2007), Yang et al. (2006)
Prenatal repeated variable stress Koenig (2006)a, Koenig et al. (2005), Lee et al. (2007)
Postnatal maternal deprivation Ellenbroek et al. (1998), Ellenbroek and Cools (2000)
Obstetric complications Cesarean section Brake et al. (1997, 2000), El-Khodor and Boksa (1998, 2000), Juarez et al. (2005), Vaillancourt and Boksa (2000)
Hypoxia/anoxia El-Khodor and Boksa (1997), Huang et al. (2009), Pereira et al. (2007), Sandager- Nielsen et al. (2004), Fendt et al. (2008)
Placental insufficiency Dieni and Rees (2003, 2005), Mallard et al. (1999), Rehn et al. (2004)
Nutritional deficiency Prenatal protein deprivation Palmer et al. (2004, 2008), Tonkiss et al. (1998), Ranade et al. (2008)
Vitamin D deficiency Eyles et al. (2009)a, Burne et al. (2004a, b), O’Loan et al. (2007)
Social isolation Postweaning social isolation rearing Fone and Porkess (2008)a, McLean et al. (2010), Powell and Geyer (2002)a, Schiavone et al. (2009)
Heuristic models
Developmental hippocampal pathology Neonatal ventral hippocampal lesion Bertrand et al. (2010), Lipska and Weinberger (2000)a, Tseng et al. (2009)a
Disruption in neuronal migration Prenatal MAM Le Pen et al. (2006), Lodge and Grace (2009)a, Moore et al. (2006)
Disruption in perinatal brain development Neonatal NMDA antagonist du Bois et al. (2008), Mouri et al. (2007)a, Nakatani-Pawlak et al. (2009), Wang et al. (2008)
a

Indicates review article; refer to the text for a more complete overview of the models

3.1 Viral and Immune-Activating Models of Schizophrenia

Epidemiological studies have linked prenatal exposure to viral and bacterial infections during early to mid-gestation with an increased risk for schizophrenia (reviewed in Brown and Susser 2002; Fatemi and Folsom 2009; Patterson 2009; but see also Selten et al. 1999a). Early studies focused on the link between influenza and schizophrenia (Mednick et al. 1988; O’Callaghan et al. 1991), but other infectious agents such as toxoplasmosis (Brown et al. 2005) and bacterial infections (Sorensen et al. 2009) have also been associated with the disease. Epidemiological findings have been corroborated by serologic evidence of gestational influenza infection during early to mid-pregnancy increasing the risk of schizophrenia threefold (Brown et al. 2004a). In addition to influenza, there is also serologic evidence of increased maternal levels of cytokines such as TNF-alpha (Buka et al. 2001) and IL-8 (Brown et al. 2004b) during pregnancy in mothers of patients with schizophrenia. Additional evidence for alterations in immune function in schizophrenia comes from the observation that higher levels of antibodies and alterations in other measures of immune function are reported in schizophrenia patients (reviewed in Patterson 2009; Schwarz et al. 2001). To examine and identify the causal relationship between the neural and behavioral consequences of prenatal exposure and immune challenges, the effects of maternal challenges with influenza virus (Shi et al. 2003), as well as other viruses [e.g., borna disease virus, lymphocytic choriomeningitis, cytomegalovirus; (Lipska and Weinberger 2000)], and immune activating agents have been investigated in animal models (for more thorough reviews, see Meyer and Feldon 2009a, b; Patterson 2009). When given at the appropriate time during gestation, these viruses or immune-activating agents can have rather selective effects on neuronal development and behavior. These prenatal immune paradigms, conducted in both rats and mice, have emerged over the last decade as some of the most important neurodevelopmental models of schizophrenia. These animal models involve exposure of pregnant rats or mice to an immune challenge with either influenza, the bacterial endotoxin lipopolysaccharide (LPS), or the viral mimic polyriboinosinic–polyribo-cytidilic acid (PolyI:C) during gestation and corresponding assessment of brain and behavioral effects in the offspring.

3.1.1 Prenatal Viral Exposure

Exposure of mice to influenza virus on gestation day 9 results in behavioral and brain abnormalities reminiscent of schizophrenia (Fatemi et al. 1998). Specifically, influenza-exposed mice showed deficits in PPI, decreased exploratory behavior, and decreased social interaction (Shi et al. 2003). Neuroanatomical abnormalities associated with in utero exposure to influenza include pyramidal cell atrophy and macrocephaly, increased glial fibrillary acidic protein (GFAP) immunoreactivity, increased glutamic acid decarboxylase (GAD) 65 and 67 proteins (Fatemi et al. 2002a, b, 2004). Prenatal exposure to influenza also results in decreased size of the lateral ventricles, disrupted corticogenesis, and reduced Reelin immunoreactivity in the frontal cortex and hippocampus (Fatemi et al. 1999).

3.1.2 Prenatal PolyI:C Exposure

Interestingly, the behavioral impairments (specifically PPI) in influenza-exposed offspring appeared to be associated with the maternal immune response and not with the viral infection per se because similar alterations in behavior were observed when the pregnant dam was treated with PolyI:C, which elicits an immune response in the mother similar to that observed with influenza (Shi et al. 2003). PolyI:C has been extensively studied in both rats and mice with varying outcomes based on the timing of exposure (Meyer and Feldon 2009b; Meyer et al. 2006b). Additional behavioral, neuropathological, and neurochemical studies further supported the prenatal PolyI:C model as a valid model of schizophrenia. Specifically, behavioral impairments in PPI, LI, reversal learning, novel object recognition, and working memory in addition to an increased sensitivity to dopamine agonists and glutamate antagonists are all observed in the offspring of mice and rats exposed to gestational PolyI:C (see also Fortier et al. 2007; Meyer et al. 2006a; Ozawa et al. 2006; Shi et al. 2003; Smith et al. 2007; Wolff and Bilkey 2008; Zuckerman et al. 2003; Zuckerman and Weiner 2003, 2005). Using MRI, a recent study showed increased lateral ventricle size and PPI deficits in mice exposed to PolyI:C in utero (Li et al. 2009). Several neuropathological studies revealed alterations in dopamine, gamma-aminobutyric acid (GABA), and glutamate systems and decreased Reelin expression in the PFC and hippocampus (reviewed in Meyer and Feldon 2009b). Alterations in dopamine neurocircuitry in the PolyI:C model include increased tyrosine hydroxylase (TH) immunoreactivity in the nucleus accumbens (NAC), decreased dopamine receptors in the prefrontal cortex (PFC), and alterations in basal and stimulated dopamine release depending on the timing of insult (Meyer et al. 2008b, c; Winter et al. 2009). Similar to the observation of decreased calcium-binding protein parvalbumin (PV) immunoreactivity in schizophrenia brain (Beasley et al. 2002; Reynolds et al. 2004), mice exposed to PolyI:C in utero have decreased PV staining in hippocampus and PFC (Meyer et al. 2008c). Increased limbic GABA-A receptor immunoreactivity in brains of PolyI:C-exposed mice further support a role for GABA in the neuropathology of the immune insult (Nyffeler et al. 2006). Prenatal PolyI:C also leads to alterations in the glutamate system, as evidenced by decreased expression of the N-methyl-D-aspartate (NMDA) receptor subunit 1 (NR1) (Meyer et al. 2008c).

3.1.3 Prenatal LPS Exposure

Administration of the bacterial endotoxin LPS to mammalian species mimics the innate immune response that is typically seen after infection with gram-negative bacteria. Hence, neurodevelopmental animal models of schizophrenia have also utilized LPS as an infectious agent during gestation. Initial studies with prenatal LPS conducted by Borrell and Romero and colleagues administered LPS every other day throughout pregnancy (Borrell et al. 2002; Romero et al. 2007). Similar to prenatal viral exposure, when pregnant rats were exposed to LPS, their offspring exhibited PPI deficits that emerged postpuberty and were reversed by administration of antipsychotics (Borrell et al. 2002; Romero et al. 2007, 2008). The offspring also showed increased TH immunoreactivity and basal dopamine levels in the NAC as well as decreased DARP-32 in frontal cortex and increased synaptophysin in hippocampus and cortex (Borrell et al. 2002; Romero et al. 2007, 2008). Subsequent studies revealed that discrete administration of LPS during gestation (embryonic days 15–16 or 18–19) disrupts PPI and increases amphetamine-induced locomotor activity in the offspring (Fortier et al. 2004, 2007). Additional CNS abnormalities in adult offspring include morphological changes in pyramidal neurons of the hippocampus and PFC (Baharnoori et al. 2009; Nolan et al. 2003).

LPS administered to pregnant rats has been shown to increase cytokines in the amniotic fluid (Urakubo et al. 2001) and in fetal plasma (Ashdown et al. 2006). Borrell et al. (Borrell et al. 2002) showed that serum levels of the cytokines IL-6 and IL-2 were significantly higher in adult offspring of LPS-infected dams. These authors suggest that elevated cytokine levels in the adult offspring may contribute to the PPI deficits observed following prenatal LPS. These data are in line with reports describing elevated levels of cytokines in the CSF and plasma of schizophrenia patients (Licinio et al. 1993; Mittleman et al. 1997).

3.1.4 Role of Cytokines in Prenatal Immune Models

Studies with PolyI:C indicated that the maternal immune response was responsible for the schizophrenia-like behavioral and neuronal effects of prenatal immune challenge but the specific elements of the immune response contributing to these impairments was not known. Subsequent studies went on to examine the role of specific cytokines in the prenatal immune models. PolyI:C administration increases both proinflammatory (IL-6, IL-1β, TNF alpha) and anti-inflammatory (IL-10) cytokines. Both IL-6 and IL-10 play an important role in mediating the effects of prenatal PolyI:C administration. For example, many of the behavioral and neuropathological effects of prenatal PolyI:C are mimicked by gestational administration of IL-6 and blocked in IL-6 knockout mice (Smith et al. 2007). Conversely, over-expression of the anti-inflammatory cytokine IL-10 blocked the emergence of behavioral abnormalities in the offspring exposed to PolyI:C in utero but lead to some behavioral abnormalities when overexpressed on its own in the absence of any immunogenic agent (Meyer et al. 2008a).

3.1.5 Neonatal Immune Activation

In addition to prenatal immune activation, other immune-based developmental models have focused on neonatal exposure of rat or mouse pups to a viral or immunogenic agent (Nawa et al. 2000). These neonatal infections are thought to model infection during the late second/early third trimester. Earlier work showed that rats with neonatal exposure to cytomegalovirus show an increased sensitivity to the PPI-disruptive effects of apomorphine (Rothschild et al. 1999). Neonatal influenza administered on postnatal day 3 or 4 resulted in PPI deficits in adult Tap1−/−(transporter associated with antigen processing 1) mice expressing reduced levels of MHC Class I (Asp et al. 2009). The PPI deficits were accompanied by increases in transcripts encoding indoleamine-pyrrole 2,3-dioxygenase (IDO) and transient increases in other enzymes in the kynurenine pathway of tryptophan metabolism and kynurenic acid (KYNA) (Asp et al. 2009; Holtze et al. 2008). These studies raise the possibility that elevations in KYNA, an endogenous NMDA antagonist and nicotinic acetylcholine (nAch) alpha-7 antagonist, may mediate in part the behavioral effects of neonatal immune activation. Neonatal exposure to PolyI:C also results in behavioral and neurochemical abnormalities later in life (Ibi et al. 2009). Again, the effects of these immune-activating agents may be mediated through increases in cytokines. Administration of cytokines (e.g., IL-1α, leukemia inhibitory factor [LIF]) to neonatal rat or mouse pups also results in locomotor hyperactivity, decreased PPI, impaired social interaction, and neuroanatomical abnormalities such as increased TH and dopamine metabolism (Tohmi et al. 2004; Tsuda et al. 2006; Watanabe et al. 2004). Similar to effects of prenatal PolyI:C treatment in mice, administration of LPS on postnatal day 7 and 9 to rat pups also produced decreased PV immunoreactivity in the hippocampus (Jenkins et al. 2009).

3.1.6 Discussion of Immune Models

Like all the studies of epidemiological risk factors, one important aspect to consider is that the risk factor of prenatal or neonatal viral infection only confers a modest increased risk for developing the disorder. Obviously, not all individuals exposed to an infection in utero subsequently develop schizophrenia. One advantage to the environmental risk factors outlined here is the possibility to study gene–environment interactions. Inducing an effect of immune activation in genetically compromised or genetically susceptible animal but not in the wildtype animal would be extremely compelling and useful to our understanding of gene–environment interactions in schizophrenia. These gene–environment interactions are beginning to be explored in relation to immune challenge models (reviewed in Ayhan et al. 2009). Examples of these gene–environment interactions include the neonatal exposure to influenza in TAP1−/− mice described above (Asp et al. 2009) and recent studies examining the effects of neonatal PolyI:C administration in DISC1 dominant-negative (DISC1 DN) mutant mice (Ibi et al. 2010). Specifically, DISC1 DN mice displayed a more pronounced response to the behavioral effects of neonatal PolyI:C compared to wildtype mice (Ibi et al. 2009). Hence, immune models are ripe for studies of gene–environment interactions and may prove useful in our understanding of the dynamic interplay between susceptibility genes and environmental risk factors.

As mentioned previously, the role of maternal behavior in the effects of prenatal immune activation need to be taken into account. In studies, in which cross-fostering was employed, mother–pup behavioral interactions accounted for some of the observed effects (Meyer et al. 2006c, 2008b). One additional consideration in studies of prenatal immune activation is that the immune challenge may produce a nutritional deficiency in the mom, which can have significant effects on the offspring (see Sect. 3.2). Dams do tend to lose weight in response to the immune-activating agent.

3.2 Maternal Malnutrition

Epidemiological studies suggest that prenatal nutritional deficiency increases the risk of developing schizophrenia (Brown and Susser 2008; Susser et al. 1996). Perhaps the most robust examination of this relationship comes from the two periods of famine, one during 1944–1945 in The Netherlands termed “The Dutch Hunger Winter” and the other during the “Chinese Famine” that took place during 1959–1961. Those offspring exposed to the famine during early gestation had a twofold increase of developing schizophrenia as adults (Susser et al. 1996; Xu et al. 2009). Candidate micronutrients that may be responsible for these abnormalities include folate, vitamin D, essential fatty acids, retinoids, and iron, all of which play a role in normal fetal brain development (reviewed in Brown and Susser 2008). In order to determine the mechanism responsible for altered brain development in response to early gestational malnutrition, several animal models have been developed: prenatal protein deficiency (or protein–calorie malnutrition) and prenatal vitamin D deficiency (reviewed in Meyer and Feldon 2009a).

3.2.1 Prenatal Protein Deficiency

Prenatal protein deficiency typically involves depriving the dam of protein prior to and during pregnancy and comparing the behavioral effects in the offspring to that of control rats that received normal levels of protein during gestation (e.g., low casein diets (6%) or adequate casein diets (25%)). Protein deprivation in rats leads to many alterations in brain development consistent with an animal model of schizophrenia including structural differences in the hippocampus, alterations in dopamine and serotonin release, and changes in glutamate receptor binding (Meyer and Feldon 2009a for review). As far as behavioral effects of prenatal protein deficiency, several alterations have been reported. Namely, female rats that underwent prenatal protein deprivation display a postpubertal emergence of PPI deficits (Palmer et al. 2004), an increased responsiveness to dopamine agonists and NMDA receptor antagonists (Palmer et al. 2008; Tonkiss et al. 1998). Impairments in working memory measured in a radial arm maze have been reported in rats exposed to prenatal protein deprivation (Ranade et al. 2008); whereas other studies have shown no difference in working memory as measured in a T-maze alternation task and in an operant delayed alternation task (Tonkiss and Galler 1990). There have yet to be any report showing reversal of the functional impairments in prenatal protein deficiency with drug treatment. Such future studies would clarify the predictive validity of the prenatal protein deficiency model of schizophrenia.

3.2.2 Prenatal Vitamin D Deficiency

Maternal vitamin D deficiency has also been examined in animal models for its role in the development of schizophrenia (Eyles et al. 2009). McGrath (1999) argues that Vitamin D deficiency may explain several risk factors for schizophrenia including maternal malnutrition, increased winter births in schizophrenia, urbanicity, and dark-skinned immigrants in cold climates. The hypothesis of vitamin D deficiency is intriguing because vitamin D is important for normal fetal brain development and deficiencies (e.g., rickets) can have profound impact on health (Eyles et al. 2009). The epidemiological evidence for this link is mixed. While maternal vitamin D supplementation reduced the risk of schizophrenia in an examination of data from a Finnish Birth Cohort (McGrath et al. 2004), other studies have supported the link only weakly (McGrath et al. 2003) or not at all (Kendell and Adams 2002; Özer et al. 2004). Nevertheless, animal models of maternal vitamin D deficiency have supported a link between the deficiency and brain and behavioral abnormalities related to schizophrenia (reviewed in Eyles et al. 2009). For example, prenatal vitamin D deficiency in rats is associated with enlarged lateral ventricles and smaller neocortical width (Eyles et al. 2003), decreased neurotrophin levels (Eyles et al. 2003; Feron et al. 2005), altered neurogenesis (Cui et al. 2007), and increased long-term potentiation (LTP) (Grecksch et al. 2009). Maternal vitamin D deficiency also produces changes in PFC, hippocampal, and NAC gene and protein expression in pathways involved in oxidative stress, synaptic plasticity, calcium homeostasis, and neurotransmission (Almeras et al. 2007; Eyles et al. 2007; McGrath et al. 2008).

Maternal vitamin D deficiency is associated with several behavioral impairments with validity for schizophrenia. For example, rats exposed to prenatal vitamin D deficiency demonstrate heightened locomotor activity in a novel environment (Burne et al. 2004a, 2006) and increased sensitivity to the NMDA antagonist MK-801 and the dopamine D2 receptor antagonist haloperidol (Kesby et al. 2006). Deficits in PPI are observed when the prenatal vitamin D deficiency is continued through the tenth postnatal week, but not when exposed in utero only (Burne et al. 2004b). Regarding cognitive tasks, rats exposed to prenatal vitamin D deficiency show impaired habituation in the hole board but no differences in spatial learning in the radial arm maze or active avoidance learning in the shuttle box (Becker et al. 2005). The normal learning abilities are not surprising, considering the reported increase in LTP (Grecksch et al. 2009). Although most studies produce vitamin D deficiency throughout gestation, there is some evidence that the detrimental effects of maternal vitamin D deficiency may be more pronounced when the deficiency occurs during the late gestational period (O’Loan et al. 2007). Initial studies of maternal vitamin D deficiency in mice suggest that while adult offspring displayed increased locomotor activity, vitamin D-deprived and control mice did not differ in PPI or social behavior (Harms et al. 2008).

3.3 Obstetric Complications

Another set of prenatal and perinatal risk factors that have been well documented are obstetric complications. As Rapoport et al. (2005) point out, the relative risk for schizophrenia-associated obstetric complications is low, with an odds ratio for the exposure to obstetric complications increasing the risk of schizophrenia estimated at 2.0 (Rapoport et al. 2005). Nevertheless, obstetric complications have been well documented and linked to schizophrenia in several independent studies. Specifically, birth complications such as pre-eclampsia, cesarean section, and perinatal hypoxia are associated with an increased risk of schizophrenia (Cannon et al. 2002; Hultman et al. 1997; Zornberg et al. 2000). These birth complications have all been modeled in animals (reviewed in Boksa 2004; Meyer and Feldon 2009a).

3.3.1 Cesarean Section

For Cesarean section (C-section), the experimental litter is removed from the uterus and kept warm until being placed with a foster mom (El-Khodor and Boksa 1997). In rats and the more precocious species, guinea pigs, C-section is associated with heightened sensitivity to the locomotor-activating effects of amphetamine and stress, deficits in PPI, and decreased and increased dopamine in the PFC and NAC, respectively (Brake et al. 1997, 2000; El-Khodor and Boksa 1998, 2000; Juarez et al. 2005; Vaillancourt and Boksa 2000). Additionally, rats born by C-section demonstrate a postpubertal increase in dopamine D1 receptor binding and increased functional response to D1 agonists (Boksa et al. 2002).

3.3.2 Perinatal Hypoxia

Both intrauterine and neonatal hypoxia have been tested in rats, mice, and guinea pigs. Both manipulations lead to widespread effects on brain morphology and neurochemistry, specifically with decreased hippocampal volume or neuronal cell loss coupled with reduced dendritic spine density and/or elongation (reviewed in Meyer and Feldon 2009a). Intrauterine hypoxia typically involves removing the intact uterus from the dam and placing it in a 37°C water bath for a certain period of time (typically around 15 min) (El-Khodor and Boksa 1997). Hence, perinatal hypoxia manipulations in practice also involve C-section. Neonatal hypoxia involves placing a pup in a chamber without oxygen or with very low levels of oxygen (e.g., 8%) for a specified period of time (Fendt et al. 2008; Nadri et al. 2007). Some methods combine the low-oxygen environment with occlusion of the carotid artery (Rice et al. 1981). The behavioral effects of intrauterine and neonatal hypoxia are mixed (reviewed in Meyer and Feldon 2009a). Whereas the near-term intrauterine hypoxia produced deficits in PPI and working memory in guinea pigs (Becker and Donnell 1952; Vaillancourt and Boksa 2000) and impairments in spatial learning in the water maze in rats(Boksa et al. 1995), no impairments in working memory as measured by spontaneous alternation in the T maze were reported in rats (Boksa et al. 1995). Rats exposed to intrauterine hypoxia did display decreased social and exploratory behavior and increased response to stress and dopamine agonists (Brake et al. 1997; reviewed in Meyer and Feldon 2009a).

Neonatal hypoxia is associated with reference and working memory impairments in the water maze and decreased hippocampal volume (Huang et al. 2009; Pereira et al. 2007). Interestingly, environmental enrichment blocked the memory impairments and hippocampal volume reduction produced by neonatal hypoxia (Pereira et al. 2007). Depending on the timing and severity of postnatal hypoxia, differing effects on PPI have been reported. Although hypoxia at postnatal day 9 altered mesolimbic dopamine neurochemistry, it did not produce differences in PPI (Sandager-Nielsen et al. 2004). Subsequent studies using subchronic exposure to hypoxia during postnatal day 4–8 did result in PPI deficits in the adult rat (Fendt et al. 2008). These animal studies of hypoxia are useful for our understanding of perinatal complications as a risk factor as they have shown many neuroanatomical abnormalities consistent with schizophrenia. The behavioral abnormalities, on the other hand, are less consistent thus making the model less useful for pharmacological intervention studies.

3.3.3 Placental Insufficiency

Placental insufficiency is another prenatal risk factor for schizophrenia (Cannon et al. 2002) which involves loss of blood flow to the developing fetus. Placental insufficiency is achieved experimentally in guinea pigs by ligation of the uterine artery and results in decreased PPI, enlargement of the lateral ventricles, reduced volume of the basal ganglia and septum, and reduced hippocampal BDNF (Dieni and Rees 2003, 2005; Mallard et al. 1999, 2000; Rehn et al. 2004). The neuroanatomical abnormalities, without any evidence of gliosis, together with the observation of deficient PPI, suggest that placental insufficiency may have face validity for schizophrenia. However, further behavioral tests and pharmacological interventions are warranted in this model (Meyer and Feldon 2009a).

3.4 Prenatal/Postnatal Stress

There is some evidence that psychological stressors during pregnancy increase the risk for schizophrenia in offspring. This association comes from the studies examining the occurrence of schizophrenia in offspring whose mothers were exposed to a stressful experience during pregnancy (reviewed in Koenig 2006; Koenig et al. 2002). Because assessing psychological stress retrospectively is very difficult, studies have focused on discrete events for the individual (e.g., death of a relative) or periods of “stress” for an entire community (e.g., war, flood). In a retrospective study of the 5-day Nazi invasion of the Netherlands in 1940, schizophrenia risk was increased when exposed to the traumatic event during the first trimester (van Os and Selten 1998). Additionally, the risk of schizophrenia was increased in offsprings whose mothers experienced the death of a relative during the first trimester pregnancy (Khashan et al. 2008). Other studies, however, have failed to report a relationship between prenatal stress and the later development of schizophrenia. For example, there was no relationship between a deadly 1953 flood in Holland and the later development of schizophrenia in exposed offspring (Selten et al. 1999b).

3.4.1 Prenatal Stress

On the basis of the association between prenatal stress and risk for schizophrenia and the observation that maternal stress may alter the programming of the fetal brain (Weinstock 2008), consequences of prenatal stress have been evaluated for their effects on behavior and neurochemical alterations associated with schizophrenia. The effects of prenatal stress on schizophrenia-related behaviors have been mixed. Rats exposed to prenatal stress show hyperactivity in a novel environment (Son et al. 2007) and an increased sensitivity to amphetamine (reviewed in Meyer and Feldon 2009a). Pups born to dams that had been restrained three times a day for 30 min during gestational days 15–22 did not exhibit deficits in PPI or LI when tested as adults (Lehmann et al. 2000). These rats actually showed slight increases in PPI, which was normalized by the combination of prenatal stress and maternal separation. Subsequent studies, however, did show impaired LI in the offspring of dams exposed to more severe stress of repeated electric foot shock. Prenatal repeated variable stress paradigm, on the other hand, produce behavioral alterations relevant to schizophrenia in the exposed offspring (Koenig 2006). Repeated variable stress described in these studies involved exposing the pregnant rat to several different stressors including 60 min restraint stress, cold exposure (4°C) for 6 h, overnight food deprivation, overcrowding during dark phase of cycle, swim stress, and lights on for 24 h. Rats are exposed to 2–3 of these stressors each day from gestation day 14 until parturition, i.e., the third week of pregnancy. Rats exposed to repeated variable stress showed impaired social interaction (Lee et al. 2007), PPI deficits, and increased sensitivity to amphetamine challenge postpuberty (Koenig et al. 2005). Interestingly, prenatally stressed rats also showed a trend toward deficits in N40 gating (Koenig et al. 2005). Thus, repeated variable stress appears to exert more robust effects on gating measures than does prenatal restraint stress.

Cognitive deficits, on the other hand, have been well documented in offspring of dams exposed to prenatal stress. Specifically, prenatal restraint stress leads to impairments in spatial learning in the water maze in rats (Lemaire et al. 2000; Szuran et al. 2000; Wu et al. 2007; Yang et al. 2006) and guinea pigs (Kapoor et al. 2009). Prenatally stressed rats also show impaired reversal learning in the water maze (Szuran et al. 2000). Additional impairments in working memory in the radial arm maze in mice (Son et al. 2006) and in the T maze delayed alternation test in rats have been associated with prenatal stress (Gue et al. 2004). Importantly, the working memory deficits in the T maze delayed alternation task were observed in prepubertal rats. The early emergence of working memory impairments is interesting to schizophrenia models because of the observation of early development of cognitive deficits in prodromal patients (Eastvold et al. 2007). Many sex differences in the behavioral effects of prenatal stress have been reported, with prenatal stress conferring more profound cognitive disruptions in males and more affective or anxiety disturbances in females (reviewed in Meyer and Feldon 2009a; Weinstock 2008). Few pharmacological studies have been conducted in the prenatal stress model, with the exception of oxytocin reversing impairments in social interaction in the model (Lee et al. 2007). Thus, further pharmacological studies should be conducted to evaluate the predictive validity of the model.

In addition to the behavioral abnormalities, prenatal stress alters brain development. Many studies report increased HPA axis activity and corresponding decreases in glucocorticoid and mineralcorticoid receptors (Szuran et al. 2000). Hippocampal abnormalities are consistently reported in mice exposed to prenatal stress. These abnormalities include decreased neurogenesis and decreased density of granule cells in the hippocampus (Lemaire et al. 2000), decreased LTP and enhanced LTD in hippocampal CA1 region (Son et al. 2006; Yang et al. 2006), and reduced NR2A and NR2B subunits of NMDA receptor in hippocampus (Son et al. 2006). Alterations in dopamine function including increased DA turnover and alterations in DA receptors and DA transporter (reviewed in Meyer and Feldon 2009a; Son et al. 2007).

In conclusion, although the prenatal stress model was developed primarily to examine factors contributing to anxiety and depressive disorders (Weinstock 2008), many of the behavioral and neurochemical abnormalities suggest that the model may prove useful for schizophrenia research (Koenig 2006).

3.4.2 Maternal Deprivation

Stress manipulations in the early postnatal period have also been assessed for their effects on schizophrenia-related behaviors. These experiments typically involve removing the pups from the dam during the first few weeks of life for various lengths of time, with shorter periods of time (3–15 min; early handling) resulting in decreased HPA axis response and longer periods of time (3 h or 24 h; maternal separation, MS) resulting in increased HPA axis activity (Meaney et al. 1991, 1993). Although early handling (i.e., brief separations) affects neuroendocrine and anxiety-related behavior, it does not appear to affect PPI (Pryce et al. 2001). Early handling increases LI when compared to nonhandled rats (Feldon et al. 1990; Weiner et al. 1985). PPI following more prolonged periods of maternal separation in rats has also been assessed (for review see Weiss and Feldon 2001). Ellenbroek et al. (1998) showed that separation from the dam for 24 h at PND 3, 6, and 9 produced deficits in PPI on PND 69 in male and female rats of the Wistar strain (Ellenbroek and Cools 2000). Maternal separation for shorter periods of time (e.g., 1–4 h/day) did not affect PPI in rats (Finamore and Port 2000; Weiss et al. 2001) or mice (Millstein et al. 2006), but did impair acoustic startle habituation (Finamore and Port 2000). These data indicate that longer periods of maternal separation (e.g., 24 h) at one point during the preweanling period may have a greater effect on PPI than shorter, repeated separations from the mother, suggesting that a certain amount of nutritional deprivation may be necessary to observe the effects of MS on PPI. Maternal separation does, however, result in cognitive deficits in adulthood. Specifically, MS rats display learning impairments in the water maze and the NORT (Aisa et al. 2007, 2008). These behavioral impairments are consistent with the observed alterations in hippocampal development (Huot et al. 2002) among other neurochemical and neuroanatomical differences (Holmes et al. 2005). In conclusion, MS produces cognitive impairments but does not consistently alter PPI. This observation, together with the lack of a clear epidemiological link between postnatal stress and schizophrenia, suggests that the MS model may not be a particularly useful model for schizophrenia research.

3.5 Postweaning Social Isolation

Social withdrawal and isolation are common features of schizophrenia that have received recent attention because of the role social factors play in the risk for schizophrenia and conversion to psychosis in prodromal patients (Addington et al. 2008). Indeed, social functioning, among other factors, predicts conversion to psychosis in patients at a high risk of developing psychosis (Cannon et al. 2008). Because of this observation, coupled with social factors contributing to the etiology of schizophrenia, we have categorized social isolation rearing as an epidemiological model in this review. When evaluating animal models of schizophrenia, one should consider both proximate and distal risk factors to the development of the disease. Thus far, we have discussed distal risk factors for the development of schizophrenia. Postweaning social isolation can be considered a model of a more proximal risk factor – social isolation.

Social isolation rearing of rodents is a developmental model relevant to schizophrenia that involves more subtle environmental manipulations leading to profound effects on behavior and brain development. Social isolation rearing of young rodents provides a nonpharmacologic method of inducing long-term alterations reminiscent of several symptoms seen in schizophrenia patients (Geyer et al. 1993; Powell and Geyer 2002). Rearing animals in social isolation is particularly consequential for species that rely on social contact after being weaned from the mother. Specifically, isolation rearing deprives rodents of social interactions during a developmental period in which play behavior emerges (Einon and Morgan 1977). Thus, as a consequence of social isolation, animals are deprived of stimuli critical to behavioral and neurobiological development (reviewed in Hall 1998). The lack of early social contact provides a model of the social isolation and social withdrawal which occurs early in the course of schizophrenia and predicts conversion to psychosis in patients at a high risk of developing psychosis (Cannon et al. 2008). Behavioral and neurochemical changes after isolation rearing in rats provide a nonlesion and nonpharmacological model to enhance our understanding of the developmentally linked emergence of neural and behavioral abnormalities in schizophrenia patients (Geyer et al. 1993; Powell and Geyer 2002).

3.5.1 Isolation Rearing: Neuroanatomical Abnormalities

Rats reared in social isolation exhibit profound abnormalities in behavior, drug responses, and neurochemistry compared to rats reared in social groups (Fone and Porkess 2008; Hall 1998; Powell and Geyer 2002). The most well documented set of studies are those that support isolation-reared rats as a model for dopamine hyper-reactivity associated with schizophrenia, such as (1) increased behavioral sensitivity to dopamine agonists (Bowling and Bardo 1994; Jones et al. 1990, 1992; Sahakian et al. 1975), (2) reduced responsivity to dopamine antagonists (Sahakian et al. 1977), (3) elevated basal and amphetamine-stimulated dopamine release in the NAC (Hall et al. 1998; Jones et al. 1992), and (4) elevated dopamine concentrations (Jones et al. 1992) and altered dopamine turnover (Blanc et al. 1980) in the frontal cortex. In addition to alterations in dopamine function, isolation-reared rats display abnormalities in the hippocampus and frontal cortex. Isolation-reared rats have increased density of 5-HT1A receptors in the hippocampus (Del-Bel et al. 2002; Preece et al. 2004). Synaptophysin is a synapse-specific protein involved in neuro-transmitter release and its expression is reduced within certain hippocampal sub-fields in schizophrenia (Eastwood and Harrison 1995). Varty et al. (1999) also reported reduced synaptophysin immunoreactivity in the dentate gyrus of isolation-reared rats. There is also evidence of reduced BDNF in the hippocampus (Scaccianoce et al. 2006) and decreased spine density in isolation-reared rats (Silva-Gomez et al. 2003b). More recent studies have pointed to further alterations in the hippocampus of isolation-reared rats. Loss of PV-positive GABA interneurons observed in isolation-reared rats (Harte et al. 2007; Schiavone et al. 2009) is very similar to that reported in the hippocampus and frontal cortex of schizophrenia patients (Reynolds et al. 2004; Reynolds and Beasley 2001). Isolation-reared rats show abnormalities in the PFC including (1) abnormal firing of pyramidal cells in the PFC upon dopamine stimulation from VTA neurons (Peters and O’Donnell 2005), (2) decreased volume of PFC (Day-Wilson et al. 2006; Schubert et al. 2009; Silva-Gomez et al. 2003b), and (3) decreased dendritic arborization in the PFC (Pascual et al. 2006; Silva-Gomez et al. 2003b).

3.5.2 Isolation Rearing: Behavioral Abnormalities

Rats reared in social isolation show deficits in PPI (Cilia et al. 2001, 2005; Geyer et al. 1993; Varty and Geyer 1998; Varty and Higgins 1995) and slow rates of startle habituation (for reviews, see Geyer et al. 1993, 2001; Powell and Geyer 2002; Weiss and Feldon 2001). More recent studies have also shown that several different strains of mice (e.g., ddY, 129T2, C57BL/6) exhibit deficits in PPI when reared in social isolation from weaning (Dai et al. 2004; Sakaue et al. 2003; Varty et al. 2006; see also Pietropaolo et al. 2008). Deficits in PPI produced by isolation rearing are developmentally specific in that they only appear when social isolation occurs early, during the postnatal period, and not in rats isolated as adults (Wilkinson et al. 1994). PPI deficits in isolation-reared rats can be reversed with both typical (Geyer et al. 1993; Varty and Higgins 1995) and atypical (Bakshi et al. 1998; Cilia et al. 2001; Varty and Higgins 1995; but see Barr et al. 2006) antipsychotic drugs. Thus, several investigators have shown predictive validity of the PPI deficits in the isolation-rearing model (summarized in Geyer et al. 2001). In addition to PPI deficits, isolated rats also exhibit abnormalities in motor activity. When tested in novel environments, isolated rats show elevated levels and slowed habituation of locomotor activity (Hall 1998; Jones et al. 1989, 1990; Lapiz et al. 2000; Paulus et al. 1998; Sahakian et al. 1975; Varty et al. 2000), increased investigatory behavior (e.g., rearings, holepokes; Lapiz et al. 2000; Paulus et al. 1998), and an increased preference for a novel environment (Hall et al. 1997). Additionally, isolation-reared rats and mice show increased anxiety-like behavior (Da Silva et al. 1996; Wright et al. 1991), deficits in fear learning (Voikar et al. 2005; Weiss et al. 2004), impaired recognition memory (e.g., novel object recognition; Bianchi et al. 2006; McLean et al. 2010; Voikar et al. 2005), reduced spatial memory (Ibi et al. 2008), and cognitive inflexibility as demonstrated by deficits in reversal learning (Krech et al. 1962; Schrijver et al. 2004) and extradimensional set-shifting tasks (McLean et al. 2010; Schrijver and Wurbel 2001). Thus, isolation rearing of rats and mice is associated with impaired sensorimotor gating, cognitive inflexibility, reductions in PFC volume and hippocampal synaptic plasticity, hyper-function of mesolimbic dopaminergic systems, and hypofunction of mesocortical dopamine, strikingly similar behavioral and neuroanatomical abnormalities as those observed in schizophrenia. Taken together, these results point towards the usefulness of the social isolation model in mimicking some behavioral, neurochemical, and neuropathological phenomena characteristic of schizophrenia.

Few studies have directly tested the mechanism by which isolation rearing exerts its effects on brain and behavioral development. Recently, a clear role for nicotinamide adenosine dinucleotide phosphate (NADPH) oxidase 2 (Nox2)-dependent oxidative mechanisms in the loss of PV interneurons and development of schizophrenia-like behavior in the isolation-rearing model was demonstrated (Schiavone et al. 2009). Corroborating our earlier work (Harte et al. 2007), Schiavone et al. (2009) found decreased PV immunoreactivity in the brains of rats reared in social isolation. This loss of PV interneurons was associated with elevations in Nox2, and the decrease in PV-staining and deficits in novel object recognition were blocked by treatment with the Nox2 inhibitor apocynin (Schiavone et al. 2009). Recent studies in mice have shown reductions in the expression of two developmental genes, Nurr1 and Npas4, in mice reared in social isolation (Ibi et al. 2008).

3.5.3 Discussion of Isolation-Rearing Model

There are several advantages to the isolation-rearing model that make it an appealing preclinical model of schizophrenia. First, the isolation-rearing model has shown a high degree of predictive validity for antipsychotic drugs. Second, there is a wealth of data on the relevant behavioral and neurochemical/neuroanatomical differences associated with postweaning social isolation, and many of these findings have been reported in both rats and mice. Finally, isolation rearing is a relatively easy procedure to conduct but does require ample housing space for individual cages. There are also several disadvantages to the model. Since the insult is ongoing, it is difficult to determine the precise timing of the effects since age is confounded with duration of isolation. Thus, there is a need for more discrete manipulations of the duration of isolation (e.g., early postnatal, pubertal). Relative to most of the other models reviewed here, isolation rearing occurs rather late in development and may thus only be relevant to early childhood and pubertal insults. Like the nVH lesion model discussed below, it can also be considered as a heuristic model to guide future studies into the pathophysiology of and treatments for schizophrenia.

4 Heuristic Neurodevelopmental Models

4.1 Neonatal Ventral Hippocampal Lesion Model

One of the first and most widely studied neurodevelopmental models of schizophrenia is the neonatal ventral hippocampal lesion (nVH) model. On the basis of the observation of developmental abnormalities in the hippocampus of schizophrenia patients, attempts to model the developmental perturbation and delayed behavioral symptomatology, similar to that of schizophrenia, have been undertaken (Lipska and Weinberger 2000; Tseng et al. 2009).

4.1.1 Neonatal Ventral Hippocampal Lesion Model: Behavioral Studies

Rats with nVH lesions show increased responsiveness (e.g., hyperlocomotion, increased stereotypy) to dopamine agonists (Lipska et al. 1993; Lipska and Weinberger 1993, 1994a; Sams-Dodd et al. 1997) and NMDA antagonists (Al-Amin et al. 2000, 2001), deficits in PPI (Francois et al. 2009a; Le Pen et al. 2003; Le Pen and Moreau 2002; Lipska et al. 1995), and more recently alterations in N40 gating (Swerdlow et al. 2007; Vohs et al. 2009). The locomotor sensitivity to stimulants and PPI deficits in rats with nVH lesions exhibit a delayed temporal pattern and do not appear until postpuberty (Lipska et al. 1993, 1995; Lipska and Weinberger 1993; but see Swerdlow et al. 2007). Additionally, nVH lesions produce social and cognitive impairments, strengthening the relationship to this model and schizophrenia symptomatology (Lipska 2004). Rats with nVH lesions display decreased social interactions, which emerge prepuberty (Becker et al. 1999; Flores et al. 2005; Sams-Dodd et al. 1997), and impaired social recognition memory (Becker and Grecksch 2000).

Cognitive deficits associated with nVH lesions encompass many of the cognitive domains deficient in schizophrenia. For example, rats sustaining nVH lesions display deficits in spatial learning in the water maze and radial arm maze (Chambers et al. 1996; Le Pen et al. 2000; Silva-Gomez et al. 2003a) as well as impaired avoidance learning (Le Pen et al. 2000) and novel object recognition. Continuous spatial delayed alternation task and discrete paired-trial variable-delay task were impaired in rats that sustained nVH lesions but not in rats sustaining adult VH lesions, lending further support to the neurodevelopmental construct validity of the nVH lesion model (Lipska et al. 2002a). Subsequent studies aimed at dissecting the relative contribution of the hippocampus and the PFC to the impairments on working memory tasks showed that nVH lesions lead to impairments in T maze delayed alternation and discrimination learning (Marquis et al. 2006). Impairments in T maze delayed alternation (Marquis et al. 2006) and in the radial arm maze (Chambers et al. 1996) were apparent at the juvenile age, suggesting early cognitive impairments in this model. Marquis et al. (2006) argue that the distinction of whether a given task is delay-dependent or not is critical to dissecting the relative contribution of mPFC and the hippocampus to working memory impairment. For example, lesions of the PFC impair performance when no delay is involved; whereas dorsal hippocampal lesions impair performance only when there are delays introduced (Winocur 1991). In several of the working memory tasks reviewed here, nVH lesions produced deficits independent of delay suggesting impaired PFC function. Additional probes of PFC function are set-shifting tasks, which probe cognitive flexibility or problem solving (Young et al. 2009). nVH lesions were associated with set-shifting deficits in the ASST (Marquis et al. 2008) and T maze set shifting (Brady 2009), with set-shifting impairments in the ASST occurring prepuberty.

Transient inactivation of the hippocampus during a critical developmental period produces many of the behavioral alterations observed in the nVH lesion model (Lipska 2004). Tetrodotoxin (TTX), which blocks voltage-gated sodium channels, infused into the VH on PND7 produced increased sensitivity to amphetamine and MK-801-induced locomotor activity in adulthood (Lipska et al. 2002b). These data suggest that the neonatal blockade during a critical period alters the development of hippocampal and related neurocircuits.

Important for the predictive validity of the nVH lesion neurodevelopmental model, some of the behavioral effects are reversed with antipsychotic drugs and putative antipsychotics. Antipsychotics blocked stress- and drug-induced hyperactivity (Al-Amin et al. 2000; Lipska and Weinberger 1994b; Rueter et al. 2004; Sams-Dodd et al. 1997) and PPI deficits (Le Pen and Moreau 2002; Rueter et al. 2004) but failed to block social impairments (Rueter et al. 2004; Sams-Dodd et al. 1997; but see Becker and Grecksch 2003) produced by nVH lesions. The nVH lesion model has also been used to test the efficacy of putative antipsychotics. For example, glycine and the glycine transporter inhibitor ORG24598 reverse PPI deficits associated with nVH lesions (Le Pen et al. 2003), and the AMPA antagonist LY293558 blocked MK-801-induced hyperactivity (Al-Amin et al. 2000). Thus, the nVH lesion model has shown predictive validity for schizophrenia pharmaco-therapy.

4.1.2 Neonatal Ventral Hippocampal Lesion Model: Neuropathological Studies

In the outset of the model, the ventral hippocampus and subiculum were targeted because of the consistent alterations in hippocampus in schizophrenia patients and because the ventral hippocampus has important connections with the PFC and NAC (Lipska 2004). Indeed, lesions of the ventral hippocampus on PND7 result in many neuropathological changes that mirror many of the brain alterations observed in schizophrenia, particularly alterations in the PFC. O’Donnell, Tseng and colleagues have suggested that reorganization occurs within the PFC following nVH lesions (reviewed in O’Donnell et al. 2002; Tseng et al. 2009). These PFC changes are indicated by a postpubertal emergence of altered dopamine–glutamate interactions (Tseng et al. 2007; Tseng and O’Donnell 2007). The inhibitory GABA interneuron system is dysregulated in response to nVH lesion. For example, several studies have shown decreased expression of GAD67 and PV in the PFC (Francois et al. 2009a; Lipska et al. 2003). Other studies, however, did not report changes in GAD67 or PV mRNA but did report abnormal responses to D2 stimulation in these interneurons (Tseng et al. 2008). While the development of a “noisy” circuit in the PFC may occur postpuberty, there is evidence of decreased dendritic spine densities in the PFC as early as PND36 (Marquis et al. 2008) and increased glucose metabolism as early as PND21 (Francois et al. 2009b). These changes in interneurons are supported by regional changes in GABAA receptor expression (Endo et al. 2007). Other neuroanatomical alterations include compromised neuronal function as evidenced by reduced NAA and glycogen synthase kinase-3β (GSK-3β), as well as reduced expression of BDNF mRNA, glutamate receptor GluR3, and glutamate transporter EAAC1 (reviewed in Lipska 2004).

4.1.3 Conclusions for nVH Lesion Model

While not modeling an epidemiological risk factor per se, the nVH model has proven extremely useful in preclinical animal studies of schizophrenia. Perhaps because it was one of the early neurodevelopmental models, there is a wealth of neuroanatomical and behavioral data on the model. While the cardinal measure of the nVH lesion model has been the postpubertal emergence of an increased sensitivity to amphetamine-induced locomotor activation, many other behavioral deficits are apparent including some that emerge prepuberty (e.g., social impairments, cognitive deficits). This prepubertal emergence of behavioral abnormalities may be very relevant to schizophrenia as early social and cognitive deficits continue to be recognized as symptoms occurring early in the progression of the disease. As reviewed here, the nVH lesion model has been used in preclinical drug development with some success. Additionally, changes in gene expression in PFC in the nVH lesion model may reveal new targets for schizophrenia genetic studies, thus offering a hypothesis-generating model for schizophrenia genetics (Wong et al. 2005). While there are many strengths to the nVH lesion model, there are also a few weaknesses. The procedure itself is technically challenging and often requires extra animals in the lesion group because some have to be excluded postmortem for improper placement or lack of a significant lesion. Many laboratories have, of course, successfully implemented the nVH lesion procedure as evidenced by the large number of publications on the topic. Additionally, recent advancements in MRI technology offer the ability to detect lesion size and regional extent in vivo, saving time and animals in a procedure that typically requires postmortem confirmation of lesion size and location (Bertrand et al. 2010). Thus, the nVH lesion model has shown face, predictive, and construct validity as an animal model of schizophrenia and is proving to be a useful tool in preclinical schizophrenia research.

4.2 Prenatal Toxin

Evidence of cytoarchitectural abnormalities in the brains of individuals with schizophrenia has led to the creation of models focused on disrupted neurogenesis (for a more complete review, see Lipska and Weinberger 2000; Lodge and Grace 2009). For example, rats exposed to the mitotic toxin methylazoxymethanol acetate (MAM) during gestation exhibit morphological abnormalities in brain regions implicated in schizophrenia (e.g., hippocampus, frontal, and entorhinal cortices; reviewed in Lodge and Grace 2009; Talamini et al. 1998, 1999) and behavioral abnormalities including deficits in PPI (Le Pen et al. 2006; Moore et al. 2006; with other reports of only modest effects, Talamini et al. 2000) and LI (Flagstad et al. 2005; Lodge et al. 2009). MAM-treated rats also show increased locomotor response to dopamine and glutamate psychostimulants (Lena et al. 2007; Moore et al. 2006) and impaired social interaction (Le Pen et al. 2006). The increased locomotor activity and PPI deficits emerged postpuberty (Le Pen et al. 2006; Moore et al. 2006). Gestational MAM also produces several cognitive deficits including impaired spatial recognition memory (Le Pen et al. 2006) and reversal learning (Flagstad et al. 2005; Moore et al. 2006). These behavioral differences produced by gestational MAM are associated with alterations in mesolimbic and mesocortical dopamine systems (Lodge and Grace 2007; Flagstad et al. 2004) and decreased PV interneuron number in the dorsolateral PFC and the hippocampus (Lodge et al. 2009; Penschuck et al. 2006). Experiments have administered MAM either during mid-gestation (E9–12) or late gestation (E17–18), with the more consistent behavioral effects occurring at the later gestational time point.

Other disruptors of neurogenesis have been evaluated for their effects on brain and behavioral development. For example, adult-onset PPI deficits are observed from disturbing neurogenesis with the antimitotic cytosine arabinocide (Ara-C) at embryonic days 19.5 and 20.5 (Elmer et al. 2004). Additionally, neonatal exposure of rats to the NOS inhibitor (L-nitroarginine) induces locomotor hypersensitivity to amphetamine and deficits in PPI (Black et al. 1999). Thus, gestational exposure to neurotoxins can have profound effects on brain development. Regarding its use as a neurodevelopmental model of schizophrenia, the MAM model has shown a certain degree of face and construct validity. Few studies, however, have tested the ability of antipsychotics or other compounds to reverse the behavioral effects. Thus, the pharmacological predictive validity of this model is yet to be determined.

4.3 Postnatal/Neonatal NMDA Antagonists

Accumulating evidence shows that perinatal NMDA-R antagonist exposures (pNM) can produce persistent behavioral and neurochemical deficits and the loss of PV interneurons (Andersen and Pouzet 2004; du Bois et al. 2008, 2009; Nakatani-Pawlak et al. 2009; Sircar and Rudy 1998; Stefani and Moghaddam 2005; Wang et al. 2008; Wiley et al. 2003). Blockade of NMDA receptors in the postnatal period leads to a range of behavioral abnormalities relevant to schizophrenia from enhancement of exploration to impaired working memory in the delayed alternation task (reviewed in Mouri et al. 2007). Perinatal NMDA receptor antagonist exposure also leads to impairments in sensorimotor gating, spatial memory, social interaction behavior, and cognitive flexibility in adulthood (Boctor and Ferguson 2009; Broberg et al. 2008, 2009; Lei et al. 2009; Mouri et al. 2007; Secher et al. 2009; Wang et al. 2003). In addition to cognitive deficits, typical of schizophrenia, rats treated postnatally with NMDA receptor antagonists also showed higher level of fear exhibited in the elevated plus maze (Wedzony et al. 2008) and impairments in conditioned fear (Hunt 2006). A decrease in the number of PV-positive cells and spine density in the frontal cortex, NAC and hippocampus was also shown in both rats (Wang et al. 2008) and mice (Nakatani-Pawlak et al. 2009) when analyzed in adulthood. Oxidative mechanisms in this model were suggested by results showing that antioxidants can prevent the appearance of behavioral disruptions in adulthood (Wang et al. 2003).

5 Discussion

Over the last two decades, several neurodevelopmental animal models of schizophrenia have emerged to assess the pathophysiology associated with schizophrenia risk factors, the consequence of early brain insult, and the efficacy of putative antipsychotics. Many of these models reviewed here have convincingly shown face, construct, and predictive validity for schizophrenia. These models support the developmental hypothesis of schizophrenia by demonstrating similar behavioral and neuropathological abnormalities to those observed in the clinical condition. Many of these models converge on several key behavioral and neuropathological abnormalities. The two most common and consistent behavioral phenotypes are deficits in sensorimotor gating as measured by PPI and increased sensitivity to the locomotor-activating effects of amphetamine. The question is then whether these behavioral abnormalities emerge out of a common pathway or are they merely the most sensitive to multiple neurocircuit abnormalities? Increased mesolimbic dopamine activity is common to most of the models which exhibit PPI deficits and increased sensitivity to amphetamine. In addition to overactive mesolimbic dopamine, one of the most consistent findings in the models is that of decreased PV immunoreactivity in either the PFC or hippocampus, indicating abnormalities in the GABAergic inhibitory circuits that are critical to normal neuronal activity. Indeed, Behrens and Sejnowski argue that these GABA interneurons are slow to develop and are particularly sensitive to early environmental insult, particularly oxidative stress (Behrens and Sejnowski 2009).

The recent progress in developing and characterizing neurodevelopmental models, combined with the progress in genetic models of schizophrenia (reviewed by Young et al. this text), offer a unique opportunity to study gene–environment interactions. Additionally, neurodevelopmental models also offer the ability to explore the “2-hit” model described by Keshavan et al. (Keshavan and Hogarty 1999) in which maldevelopment during two critical time periods, early brain development and then adolescence, may lead to the development of schizophrenia. While animal models offer us the ability to probe in more depth the underlying pathology associated with an early environmental manipulation, in vivo animal imaging techniques such as MRI are emerging as a useful tool to evaluate structural changes in brain in the developing animal. Initial studies using MRI have been applied to social isolation rearing (Schubert et al. 2009), nVHs (Bertrand et al. 2010), and prenatal PolyI:C (Li et al. 2009; Piontkewitz et al. 2009). Future neurodevelopmental studies may benefit from this approach which allows for the determination of the ontogeny of structural abnormalities produced by the developmental manipulations.

Findings from neurodevelopmental animal models may also aid in our understanding of the processes leading to the development of schizophrenia during the prodromal phase of the disease and inform the debate on prophylactic treatments aimed at thwarting the progression to psychosis (Powell et al. 2003). Hence, several exciting pharmacological intervention studies have been conducted in which both antipsychotics and antidepressants have shown efficacy as preventive treatments in several neurodevelopmental models (Meyer et al. 2008d; Piontkewitz et al. 2009; Richtand et al. 2006). Specifically, risperidone administered from PND35–56 prevented amphetamine-induced locomotor sensitivity in the nVH lesion model (Richtand et al. 2006). In the PolyI:C model, periadolescent clozapine and fluoxetine (PND35–65) blocked the emergence of schizophrenia-like behavioral profile (PPI, LI, amphetamine-induced hyperlocomotion) in mice (Meyer et al. 2008d), and adolescent (PND34–47) administration of clozapine blocked LI deficits and amphetamine-induced hyperlocomotion in PolyI:C-exposed rats (Piontkewitz et al. 2009). Interestingly, the structural abnormalities observed with prenatal PolyI:C, enlarged lateral ventricles and reduced hippocampal volume as measured by MRI, were also prevented with adolescent clozapine treatment in this same study (Piontkewitz et al. 2009). Thus, neurodevelopmental models may aid in the debate on the efficacy and safety of early preventive treatments during the prodromal phase of illness.

As alluded to in the introductory sections, there are several experimental considerations to take into account when conducting developmental studies such as these, with the two primary concerns being cross-fostering and litter effects. Cross-fostering can be done in one of two ways. One method for determining the effect of the prenatal manipulation on maternal behavior is to give dams of each treatment group litters of both control and prenatally exposed neonates. The other method would be to use control lactating surrogate dams that had not been exposed to either treatment. Indeed, there is evidence in some of the neurodevelopmental models reviewed here that the mother–pup behavioral interaction accounts for some of the observed effects (Meyer et al. 2006c, 2008b). Litter effects can pose statistical problems for the analysis because of the interdependence of individual animals from one litter. Often, the entire litter is used and each animal is treated as an independent observation in the ANOVA. In order to avoid this problem of artificially inflating the sample size, one common practice is to sample only one animal from each litter for the experimental analyses. As Zorilla (Zorrilla 1997) argues, using a representative animal from each litter (or “two-stage sampling”) also poses problems for statistical analyses when the within litter variability on a given measure is high. One solution is to average at least two pups per litter, with additional observations not necessarily adding much more power (Zorrilla 1997). Another option is to try to minimize litter effects in the design by using a stud for multiple dams assigned to different treatment groups or using inbred strains (Zorrilla 1997). One easy approach for postnatal manipulations is to randomize treatment conditions within litter. In addition to these experimental design considerations, the effect of litter can be handled somewhat at the analysis step. Covarying for litter in the ANOVA is an option, but it requires at least two littermate observations for each between-litter effect (i.e., two littermates per sex per treatment condition). Perhaps the best way to deal with litter effects is to combine the experimental design suggestions with a within litter statistical analysis in which litter is nested within the treatment condition (reviewed in Zorrilla 1997).

In conclusion, developmental models focused on epidemiological risk factors and neurodevelopmental anomalies have contributed to our understanding of the developing brain, the neuropathology of schizophrenia, and treatment approaches for this debilitating disease. Future studies in this area should continue to examine the 2-hit hypothesis of schizophrenia through the combination of genetic and environmental manipulations and early plus late environmental manipulations. The manipulations used to study secondary or “late hits” could be based on recent findings of putative risk factors that may increase the conversion to psychosis in high-risk individuals (Cannon et al. 2008), many of which occur during adolescence (e.g., social isolation, substance abuse, etc.). Lastly, these models are uniquely suited for epigenetic studies aimed at determining the mechanism by which these manipulations exert long-term effects on brain and behavioral development.

Abbreviations

ASST

Attentional set-shifting task

DA

Dopamine

GABA

Gamma-aminobutyric acid

IL-1α

Interleukin-1alpha

IL-6

Interleukin-6

LI

Latent inhibition

LPS

Lipopolysaccharide

NMDA

N-methyl-D-aspartate

NORT

Novel object recognition

Nox2

NADPH oxidase 2

nVH

Neonatal ventral hippocampal lesion

PCP

Phencyclidine

PFC

Prefrontal cortex

PND

Postnatal day

pNM

Perinatal NMDA antagonist

PolyI:C

Polyriboinosinic–polyribocytidilic acid

PPI

Prepulse inhibition

PV

Parvalbumin

TH

Tyrosine hydroxylase

References

  1. Addington J, Addington D, Gasbarre L. Distractibility and symptoms in schizophrenia. J Psychiatry Neurosci. 1997;22:180–184. [PMC free article] [PubMed] [Google Scholar]
  2. Addington J, Penn D, Woods SW, Addington D, Perkins DO. Social functioning in individuals at clinical high risk for psychosis. Schizophr Res. 2008;99:119–124. doi: 10.1016/j.schres.2007.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Aisa B, Tordera R, Lasheras B, Del Rio J, Ramirez MJ. Cognitive impairment associated to HPA axis hyperactivity after maternal separation in rats. Psychoneuroendocrinology. 2007;32:256–266. doi: 10.1016/j.psyneuen.2006.12.013. [DOI] [PubMed] [Google Scholar]
  4. Aisa B, Tordera R, Lasheras B, Del Rio J, Ramirez MJ. Effects of maternal separation on hypothalamic-pituitary-adrenal responses, cognition and vulnerability to stress in adult female rats. Neuroscience. 2008;154:1218–1226. doi: 10.1016/j.neuroscience.2008.05.011. [DOI] [PubMed] [Google Scholar]
  5. Al-Amin HA, Weinberger DR, Lipska BK. Exaggerated MK-801-induced motor hyperactivity in rats with the neonatal lesion of the ventral hippocampus. Behav Pharmacol. 2000;11:269–278. doi: 10.1097/00008877-200006000-00010. [DOI] [PubMed] [Google Scholar]
  6. Al-Amin HA, Shannon Weickert C, Weinberger DR, Lipska BK. Delayed onset of enhanced MK-801-induced motor hyperactivity after neonatal lesions of the rat ventral hippocampus. Biol Psychiatry. 2001;49:528–539. doi: 10.1016/s0006-3223(00)00968-9. [DOI] [PubMed] [Google Scholar]
  7. Almeras L, Eyles D, Benech P, Laffite D, Villard C, Patatian A, Boucraut J, Mackay-Sim A, McGrath J, Feron F. Developmental vitamin D deficiency alters brain protein expression in the adult rat: implications for neuropsychiatric disorders. Proteomics. 2007;7:769–780. doi: 10.1002/pmic.200600392. [DOI] [PubMed] [Google Scholar]
  8. Andersen JD, Pouzet B. Spatial memory deficits induced by perinatal treatment of rats with PCP and reversal effect of D-serine. Neuropsychopharmacology. 2004;29:1080–1090. doi: 10.1038/sj.npp.1300394. [DOI] [PubMed] [Google Scholar]
  9. Arnold SE, Hyman BT, Van Hoesen GW, Damasio AR. Some cytoarchitectural abnormalities of the entorhinal cortex in schizophrenia. Arch Gen Psychiatry. 1991;48:625–632. doi: 10.1001/archpsyc.1991.01810310043008. [DOI] [PubMed] [Google Scholar]
  10. Arnold SE, Ruscheinsky DD, Han LY. Further evidence of abnormal cytoarchitecture of the entorhinal cortex in schizophrenia using spatial point pattern analyses. Biol Psychiatry. 1997;42:639–647. doi: 10.1016/s0006-3223(97)00142-x. [DOI] [PubMed] [Google Scholar]
  11. Ashdown H, Dumont Y, Ng M, Poole S, Boksa P, Luheshi GN. The role of cytokines in mediating effects of prenatal infection on the fetus: implications for schizophrenia. Mol Psychiatry. 2006;11:47–55. doi: 10.1038/sj.mp.4001748. [DOI] [PubMed] [Google Scholar]
  12. Asp L, Holtze M, Powell SB, Karlsson H, Erhardt S. Neonatal infection with neurotropic influenza A virus induces the kynurenine pathway in early life and disrupts sensorimotor gating in adult Tap1−/− mice. Int J Neuropsychopharmacol. 2009;17:1–11. doi: 10.1017/S1461145709990253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Ayhan Y, Sawa A, Ross CA, Pletnikov MV. Animal models of gene–environment interactions in schizophrenia. Behav Brain Res. 2009;204:274–281. doi: 10.1016/j.bbr.2009.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Baharnoori M, Brake WG, Srivastava LK. Prenatal immune challenge induces developmental changes in the morphology of pyramidal neurons of the prefrontal cortex and hippocampus in rats. Schizophr Res. 2009;107:99–109. doi: 10.1016/j.schres.2008.10.003. [DOI] [PubMed] [Google Scholar]
  15. Bakshi VP, Swerdlow NR, Braff DL, Geyer MA. Reversal of isolation rearing-induced deficits in prepulse inhibition by Seroquel and olanzapine. Biol Psychiatry. 1998;43:436–445. doi: 10.1016/s0006-3223(97)00246-1. [DOI] [PubMed] [Google Scholar]
  16. Barr AM, Powell SB, Markou A, Geyer MA. Iloperidone reduces sensorimotor gating deficits in pharmacological models, but not a developmental model, of disrupted prepulse inhibition in rats. Neuropharmacology. 2006;51:457–465. doi: 10.1016/j.neuropharm.2006.04.004. [DOI] [PubMed] [Google Scholar]
  17. Beasley CL, Zhang ZJ, Patten I, Reynolds GP. Selective deficits in prefrontal cortical GABAergic neurons in schizophrenia defined by the presence of calcium-binding proteins. Biol Psychiatry. 2002;52:708–715. doi: 10.1016/s0006-3223(02)01360-4. [DOI] [PubMed] [Google Scholar]
  18. Becker RF, Donnell W. Learning behavior in guinea pigs subjected to asphyxia at birth. J Comp Physiol Psychol. 1952;45:153–162. doi: 10.1037/h0055846. [DOI] [PubMed] [Google Scholar]
  19. Becker A, Grecksch G. Social memory is impaired in neonatally ibotenic acid lesioned rats. Behav Brain Res. 2000;109:137–140. doi: 10.1016/s0166-4328(99)00163-1. [DOI] [PubMed] [Google Scholar]
  20. Becker A, Grecksch G. Haloperidol and clozapine affect social behaviour in rats postnatally lesioned in the ventral hippocampus. Pharmacol Biochem Behav. 2003;76:1–8. doi: 10.1016/s0091-3057(03)00139-4. [DOI] [PubMed] [Google Scholar]
  21. Becker A, Grecksch G, Bernstein HG, Hollt V, Bogerts B. Social behaviour in rats lesioned with ibotenic acid in the hippocampus: quantitative and qualitative analysis. Psychopharmacology (Berl) 1999;144:333–338. doi: 10.1007/s002130051015. [DOI] [PubMed] [Google Scholar]
  22. Becker A, Eyles DW, McGrath JJ, Grecksch G. Transient prenatal vitamin D deficiency is associated with subtle alterations in learning and memory functions in adult rats. Behav Brain Res. 2005;161:306–312. doi: 10.1016/j.bbr.2005.02.015. [DOI] [PubMed] [Google Scholar]
  23. Behrens MM, Sejnowski TJ. Does schizophrenia arise from oxidative dysregulation of parvalbumin-interneurons in the developing cortex? Neuropharmacology. 2009;57:193–200. doi: 10.1016/j.neuropharm.2009.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Bertolino A, Weinberger DR. Proton magnetic resonance spectroscopy in schizophrenia. Eur J Radiol. 1999;30:132–141. doi: 10.1016/s0720-048x(99)00052-2. [DOI] [PubMed] [Google Scholar]
  25. Bertrand JB, Langlois JB, Begou M, Volle J, Brun P, d’Amato T, Saoud M, Suaud-Chagny MF. Longitudinal MRI monitoring of brain damage in the neonatal ventral hippocampal lesion rat model of schizophrenia. Hippocampus. 2010;20:264–278. doi: 10.1002/hipo.20628. [DOI] [PubMed] [Google Scholar]
  26. Bianchi M, Fone KF, Azmi N, Heidbreder CA, Hagan JJ, Marsden CA. Isolation rearing induces recognition memory deficits accompanied by cytoskeletal alterations in rat hippocampus. Eur J Neurosci. 2006;24:2894–2902. doi: 10.1111/j.1460-9568.2006.05170.x. [DOI] [PubMed] [Google Scholar]
  27. Black MD, Selk DE, Hitchcock JM, Wettstein JG, Sorensen SM. On the effect of neonatal nitric oxide synthase inhibition in rats: a potential neurodevelopmental model of schizophrenia. Neuropharmacology. 1999;38:1299–1306. doi: 10.1016/s0028-3908(99)00041-6. [DOI] [PubMed] [Google Scholar]
  28. Blanc G, Herve D, Simon H, Lisoprawski A, Glowinski J, Tassin JP. Response to stress of mesocortico-frontal dopaminergic neurones in rats after long-term isolation. Nature. 1980;284:265–267. doi: 10.1038/284265a0. [DOI] [PubMed] [Google Scholar]
  29. Boctor SY, Ferguson SA. Neonatal NMDA receptor antagonist treatments have no effects on prepulse inhibition of postnatal day 25 Sprague-Dawley rats. Neurotoxicology. 2009;30:151–154. doi: 10.1016/j.neuro.2008.10.011. [DOI] [PubMed] [Google Scholar]
  30. Boksa P. Animal models of obstetric complications in relation to schizophrenia. Brain Res Brain Res Rev. 2004;45:1–17. doi: 10.1016/j.brainresrev.2004.01.001. [DOI] [PubMed] [Google Scholar]
  31. Boksa P, El-Khodor BF. Birth insult interacts with stress at adulthood to alter dopaminergic function in animal models: possible implications for schizophrenia and other disorders. Neurosci Biobehav Rev. 2003;27:91–101. doi: 10.1016/s0149-7634(03)00012-5. [DOI] [PubMed] [Google Scholar]
  32. Boksa P, Krishnamurthy A, Brooks W. Effects of a period of asphyxia during birth on spatial learning in the rat. Pediatr Res. 1995;37:489–496. doi: 10.1203/00006450-199504000-00018. [DOI] [PubMed] [Google Scholar]
  33. Boksa P, Zhang Y, Bestawros A. Dopamine D1 receptor changes due to caesarean section birth: effects of anesthesia, developmental time course, and functional consequences. Exp Neurol. 2002;175:388–397. doi: 10.1006/exnr.2002.7896. [DOI] [PubMed] [Google Scholar]
  34. Borrell J, Vela JM, Arevalo-Martin A, Molina-Holgado E, Guaza C. Prenatal immune challenge disrupts sensorimotor gating in adult rats. Implications for the etiopathogenesis of schizophrenia. Neuropsychopharmacology. 2002;26:204–215. doi: 10.1016/S0893-133X(01)00360-8. [DOI] [PubMed] [Google Scholar]
  35. Bowling SL, Bardo MT. Locomotor and rewarding effects of amphetamine in enriched, social, and isolate reared rats. Pharmacol Biochem Behav. 1994;48:459–464. doi: 10.1016/0091-3057(94)90553-3. [DOI] [PubMed] [Google Scholar]
  36. Boyd JH, Pulver AE, Stewart W. Season of birth: schizophrenia and bipolar disorder. Schizophr Bull. 1986;12:173–186. doi: 10.1093/schbul/12.2.173. [DOI] [PubMed] [Google Scholar]
  37. Brady AM. Neonatal ventral hippocampal lesions disrupt set-shifting ability in adult rats. Behav Brain Res. 2009;205:294–298. doi: 10.1016/j.bbr.2009.07.025. [DOI] [PubMed] [Google Scholar]
  38. Braff DL. Prepulse inhibition (PPI) of the startle reflex: A window on the brain in schizophrenia. Behavioral Neurobiology of Schizophrenia and Its Treatment. In: Swerdlow N, editor. Curr Topics in Behav Neurosci. Springer; Heidelberg: 2010. [DOI] [PubMed] [Google Scholar]
  39. Braff DL, Geyer MA, Swerdlow NR. Human studies of prepulse inhibition of startle: normal subjects, patient groups, and pharmacological studies. Psychopharmacology (Berl) 2001;156:234–258. doi: 10.1007/s002130100810. [DOI] [PubMed] [Google Scholar]
  40. Brake WG, Boksa P, Gratton A. Effects of perinatal anoxia on the acute locomotor response to repeated amphetamine administration in adult rats. Psychopharmacology (Berl) 1997;133:389–395. doi: 10.1007/s002130050419. [DOI] [PubMed] [Google Scholar]
  41. Brake WG, Sullivan RM, Gratton A. Perinatal distress leads to lateralized medial prefrontal cortical dopamine hypofunction in adult rats. J Neurosci. 2000;20:5538–5543. doi: 10.1523/JNEUROSCI.20-14-05538.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Broberg BV, Dias R, Glenthoj BY, Olsen CK. Evaluation of a neurodevelopmental model of schizophrenia – early postnatal PCP treatment in attentional set-shifting. Behav Brain Res. 2008;190:160–163. doi: 10.1016/j.bbr.2008.02.020. [DOI] [PubMed] [Google Scholar]
  43. Broberg BV, Glenthoj BY, Dias R, Larsen DB, Olsen CK. Reversal of cognitive deficits by an ampakine (CX516) and sertindole in two animal models of schizophrenia – sub-chronic and early postnatal PCP treatment in attentional set-shifting. Psychopharmacology (Berl) 2009;206:631–640. doi: 10.1007/s00213-009-1540-5. [DOI] [PubMed] [Google Scholar]
  44. Brown AS, Susser ES. In utero infection and adult schizophrenia. Ment Retard Dev Disabil Res Rev. 2002;8:51–57. doi: 10.1002/mrdd.10004. [DOI] [PubMed] [Google Scholar]
  45. Brown AS, Susser ES. Prenatal nutritional deficiency and risk of adult schizophrenia. Schizophr Bull. 2008;34:1054–1063. doi: 10.1093/schbul/sbn096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. 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. 2004a;61:774–780. doi: 10.1001/archpsyc.61.8.774. [DOI] [PubMed] [Google Scholar]
  47. Brown AS, Hooton J, Schaefer CA, Zhang H, Petkova E, Babulas V, Perrin M, Gorman JM, Susser ES. Elevated maternal interleukin-8 levels and risk of schizophrenia in adult offspring. Am J Psychiatry. 2004b;161:889–895. doi: 10.1176/appi.ajp.161.5.889. [DOI] [PubMed] [Google Scholar]
  48. 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. doi: 10.1176/appi.ajp.162.4.767. [DOI] [PubMed] [Google Scholar]
  49. Bruins Slot LA, Kleven MS, Newman-Tancredi A. Effects of novel antipsychotics with mixed D(2) antagonist/5-HT(1A) agonist properties on PCP-induced social interaction deficits in the rat. Neuropharmacology. 2005;49:996–1006. doi: 10.1016/j.neuropharm.2005.05.013. [DOI] [PubMed] [Google Scholar]
  50. Buka SL, Tsuang MT, Torrey EF, Klebanoff MA, Wagner RL, Yolken RH. Maternal cytokine levels during pregnancy and adult psychosis. Brain Behav Immun. 2001;15:411–420. doi: 10.1006/brbi.2001.0644. [DOI] [PubMed] [Google Scholar]
  51. Burne TH, Becker A, Brown J, Eyles DW, Mackay-Sim A, McGrath JJ. Transient prenatal vitamin D deficiency is associated with hyperlocomotion in adult rats. Behav Brain Res. 2004a;154:549–555. doi: 10.1016/j.bbr.2004.03.023. [DOI] [PubMed] [Google Scholar]
  52. Burne TH, Feron F, Brown J, Eyles DW, McGrath JJ, Mackay-Sim A. Combined prenatal and chronic postnatal vitamin D deficiency in rats impairs prepulse inhibition of acoustic startle. Physiol Behav. 2004b;81:651–655. doi: 10.1016/j.physbeh.2004.03.004. [DOI] [PubMed] [Google Scholar]
  53. Burne TH, O’Loan J, McGrath JJ, Eyles DW. Hyperlocomotion associated with transient prenatal vitamin D deficiency is ameliorated by acute restraint. Behav Brain Res. 2006;174:119–124. doi: 10.1016/j.bbr.2006.07.015. [DOI] [PubMed] [Google Scholar]
  54. Cannon M, Jones PB, Murray RM. Obstetric complications and schizophrenia: historical and meta-analytic review. Am J Psychiatry. 2002;159:1080–1092. doi: 10.1176/appi.ajp.159.7.1080. [DOI] [PubMed] [Google Scholar]
  55. Cannon TD, van Erp TGM, Bearden CE, Loewy R, Thompson P, Toga AW, Huttunnen MO, Keshavan MS, Seidman LJ, Tsuang MT. Early and late neurodevelopmental influences in the prodrome to schizophrenia: contributions of genes, environment, and their interactions. Schizophr Bull. 2003;29:653–669. doi: 10.1093/oxfordjournals.schbul.a007037. [DOI] [PubMed] [Google Scholar]
  56. Cannon TD, Hennah W, van Erp TG, Thompson PM, Lonnqvist J, Huttunen M, Gasperoni T, Tuulio-Henriksson A, Pirkola T, Toga AW, Kaprio J, Mazziotta J, Peltonen L. Association of DISC1/TRAX haplotypes with schizophrenia, reduced prefrontal gray matter, and impaired short- and long-term memory. Arch Gen Psychiatry. 2005;62:1205–1213. doi: 10.1001/archpsyc.62.11.1205. [DOI] [PubMed] [Google Scholar]
  57. Cannon TD, Cadenhead K, Cornblatt B, Woods SW, Addington J, Walker E, Seidman LJ, Perkins D, Tsuang M, McGlashan T, Heinssen R. Prediction of psychosis in youth at high clinical risk: a multisite longitudinal study in North America. Arch Gen Psychiatry. 2008;65:28–37. doi: 10.1001/archgenpsychiatry.2007.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Chambers RA, Moore J, McEvoy JP, Levin ED. Cognitive effects of neonatal hippocampal lesions in a rat model of schizophrenia. Neuropsychopharmacology. 1996;15:587–594. doi: 10.1016/S0893-133X(96)00132-7. [DOI] [PubMed] [Google Scholar]
  59. Chudasama Y, Robbins TW. Psychopharmacological approaches to modulating attention in the five-choice serial reaction time task: implications for schizophrenia. Psychopharmacology (Berl) 2004;174:86–98. doi: 10.1007/s00213-004-1805-y. [DOI] [PubMed] [Google Scholar]
  60. Cilia J, Reavill C, Hagan JJ, Jones DN. Long-term evaluation of isolation-rearing induced prepulse inhibition deficits in rats. Psychopharmacology (Berl) 2001;156:327–337. doi: 10.1007/s002130100786. [DOI] [PubMed] [Google Scholar]
  61. Cilia J, Hatcher PD, Reavill C, Jones DN. Long-term evaluation of isolation-rearing induced prepulse inhibition deficits in rats: an update. Psychopharmacology (Berl) 2005;180:57–62. doi: 10.1007/s00213-004-2139-5. [DOI] [PubMed] [Google Scholar]
  62. Clancy B, Darlington RB, Finlay BL. Translating developmental time across mammalian species. Neuroscience. 2001;105:7–17. doi: 10.1016/s0306-4522(01)00171-3. [DOI] [PubMed] [Google Scholar]
  63. Clancy B, Finlay BL, Darlington RB, Anand KJ. Extrapolating brain development from experimental species to humans. Neurotoxicology. 2007;28:931–937. doi: 10.1016/j.neuro.2007.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Crawley JN. Mouse behavioral assays relevant to the symptoms of autism. Brain Pathol. 2007;17:448–459. doi: 10.1111/j.1750-3639.2007.00096.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Cui X, McGrath JJ, Burne TH, Mackay-Sim A, Eyles DW. Maternal vitamin D depletion alters neurogenesis in the developing rat brain. Int J Dev Neurosci. 2007;25:227–232. doi: 10.1016/j.ijdevneu.2007.03.006. [DOI] [PubMed] [Google Scholar]
  66. Da Silva NL, Ferreira VM, Carobrez Ade P, Morato GS. Individual housing from rearing modifies the performance of young rats on the elevated plus-maze apparatus. Physiol Behav. 1996;60:1391–1396. doi: 10.1016/s0031-9384(96)00254-5. [DOI] [PubMed] [Google Scholar]
  67. Dai H, Okuda H, Iwabuchi K, Sakurai E, Chen Z, Kato M, Iinuma K, Yanai K. Social isolation stress significantly enhanced the disruption of prepulse inhibition in mice repeatedly treated with methamphetamine. Ann N Y Acad Sci. 2004;1025:257–266. doi: 10.1196/annals.1316.032. [DOI] [PubMed] [Google Scholar]
  68. Day-Wilson KM, Jones DNC, Southam E, Cilia J, Totterdell S. Medial prefrontal cortex volume loss in rats with isolation rearing-induced deficits in prepulse inhibition of acoustic startle. Neuroscience. 2006;141:1113–1121. doi: 10.1016/j.neuroscience.2006.04.048. [DOI] [PubMed] [Google Scholar]
  69. Dean K, Bramon E, Murray RM. The causes of schizophrenia: neurodevelopment and other risk factors. J Psychiatr Pract. 2003;9:442–454. doi: 10.1097/00131746-200311000-00007. [DOI] [PubMed] [Google Scholar]
  70. Del-Bel EA, Joca SR, Padovan CM, Guimaraes FS. Effects of isolation-rearing on serotonin-1A and M1-muscarinic receptor messenger RNA expression in the hipocampal formation of rats. Neurosci Lett. 2002;332:123–126. doi: 10.1016/s0304-3940(02)00933-3. [DOI] [PubMed] [Google Scholar]
  71. Dieni S, Rees S. Dendritic morphology is altered in hippocampal neurons following prenatal compromise. J Neurobiol. 2003;55:41–52. doi: 10.1002/neu.10194. [DOI] [PubMed] [Google Scholar]
  72. Dieni S, Rees S. BDNF and TrkB protein expression is altered in the fetal hippocampus but not cerebellum after chronic prenatal compromise. Exp Neurol. 2005;192:265–273. doi: 10.1016/j.expneurol.2004.06.003. [DOI] [PubMed] [Google Scholar]
  73. du Bois TM, Huang XF, Deng C. Perinatal administration of PCP alters adult behaviour in female Sprague-Dawley rats. Behav Brain Res. 2008;188:416–419. doi: 10.1016/j.bbr.2007.11.017. [DOI] [PubMed] [Google Scholar]
  74. du Bois TM, Deng C, Han M, Newell KA, Huang XF. Excitatory and inhibitory neuro-transmission is chronically altered following perinatal NMDA receptor blockade. Eur Neuropsychopharmacol. 2009;19:256–265. doi: 10.1016/j.euroneuro.2008.12.002. [DOI] [PubMed] [Google Scholar]
  75. Eastvold AD, Heaton RK, Cadenhead KS. Neurocognitive deficits in the (putative) pro-drome and first episode of psychosis. Schizophr Res. 2007;93:266–277. doi: 10.1016/j.schres.2007.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Eastwood SL, Harrison PJ. Decreased synaptophysin in the medial temporal lobe in schizophrenia demonstrated using immunoautoradiography. Neuroscience. 1995;69:339–343. doi: 10.1016/0306-4522(95)00324-c. [DOI] [PubMed] [Google Scholar]
  77. Einon DF, Morgan MJ. A critical period for social isolation in the rat. Dev Psychobiol. 1977;10:123–132. doi: 10.1002/dev.420100205. [DOI] [PubMed] [Google Scholar]
  78. El-Khodor BF, Boksa P. Long-term reciprocal changes in dopamine levels in prefrontal cortex versus nucleus accumbens in rats born by Caesarean section compared to vaginal birth. Exp Neurol. 1997;145:118–129. doi: 10.1006/exnr.1997.6437. [DOI] [PubMed] [Google Scholar]
  79. El-Khodor BF, Boksa P. Birth insult increases amphetamine-induced behavioral responses in the adult rat. Neuroscience. 1998;87:893–904. doi: 10.1016/s0306-4522(98)00194-8. [DOI] [PubMed] [Google Scholar]
  80. El-Khodor BF, Boksa P. Transient birth hypoxia increases behavioral responses to repeated stress in the adult rat. Behav Brain Res. 2000;107:171–175. doi: 10.1016/s0166-4328(99)00119-9. [DOI] [PubMed] [Google Scholar]
  81. Ellenbroek BA, Cools AR. The long-term effects of maternal deprivation depend on the genetic background. Neuropsychopharmacology. 2000;23:99–106. doi: 10.1016/S0893-133X(00)00088-9. [DOI] [PubMed] [Google Scholar]
  82. Ellenbroek BA, van den Kroonenberg PT, Cools AR. The effects of an early stressful life event on sensorimotor gating in adult rats. Schizophr Res. 1998;30:251–260. doi: 10.1016/s0920-9964(97)00149-7. [DOI] [PubMed] [Google Scholar]
  83. Elmer GI, Sydnor J, Guard H, Hercher E, Vogel MW. Altered prepulse inhibition in rats treated prenatally with the antimitotic Ara-C: an animal model for sensorimotor gating deficits in schizophrenia. Psychopharmacology. 2004;174:177–189. doi: 10.1007/s00213-003-1757-7. [DOI] [PubMed] [Google Scholar]
  84. Endo K, Hori T, Abe S, Asada T. Alterations in GABA(A) receptor expression in neonatal ventral hippocampal lesioned rats: comparison of prepubertal and postpubertal periods. Synapse. 2007;61:357–366. doi: 10.1002/syn.20393. [DOI] [PubMed] [Google Scholar]
  85. Engelmann M, Wotjak CT, Landgraf R. Social discrimination procedure: an alternative method to investigate juvenile recognition abilities in rats. Physiol Behav. 1995;58:315–321. doi: 10.1016/0031-9384(95)00053-l. [DOI] [PubMed] [Google Scholar]
  86. Eyles D, Brown J, Mackay-Sim A, McGrath J, Feron F. Vitamin D3 and brain development. Neuroscience. 2003;118:641–653. doi: 10.1016/s0306-4522(03)00040-x. [DOI] [PubMed] [Google Scholar]
  87. Eyles D, Almeras L, Benech P, Patatian A, Mackay-Sim A, McGrath J, Feron F. Developmental vitamin D deficiency alters the expression of genes encoding mitochondrial, cytoskeletal and synaptic proteins in the adult rat brain. J Steroid Biochem Mol Biol. 2007;103:538–545. doi: 10.1016/j.jsbmb.2006.12.096. [DOI] [PubMed] [Google Scholar]
  88. Eyles DW, Feron F, Cui X, Kesby JP, Harms LH, Ko P, McGrath JJ, Burne TH. Developmental vitamin D deficiency causes abnormal brain development. Psychoneuroendocrinology. 2009;34(Suppl 1):S247–S257. doi: 10.1016/j.psyneuen.2009.04.015. [DOI] [PubMed] [Google Scholar]
  89. Falkai P, Schneider-Axmann T, Honer WG. Entorhinal cortex pre-alpha cell clusters in schizophrenia: quantitative evidence of a developmental abnormality. Biol Psychiatry. 2000;47:937–943. doi: 10.1016/s0006-3223(99)00250-4. [DOI] [PubMed] [Google Scholar]
  90. Fatemi SH, Folsom TD. The neurodevelopmental hypothesis of schizophrenia, revisited. Schizophr Bull. 2009;35:528–548. doi: 10.1093/schbul/sbn187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Fatemi SH, Sidwell R, Akhter P, Sedgewick J, Thuras P, Bailey K, Kist D. Human influenza viral infection in utero increases nNOS expression in hippocampi of neonatal mice. Synapse. 1998;29:84–88. doi: 10.1002/(SICI)1098-2396(199805)29:1<84::AID-SYN8>3.0.CO;2-7. [DOI] [PubMed] [Google Scholar]
  92. Fatemi SH, Emamian ES, Kist D, Sidwell RW, Nakajima K, Akhter P, Shier A, Sheikh S, Bailey K. Defective corticogenesis and reduction in Reelin immunoreactivity in cortex and hippocampus of prenatally infected neonatal mice. Mol Psychiatry. 1999;4:145–154. doi: 10.1038/sj.mp.4000520. [DOI] [PubMed] [Google Scholar]
  93. Fatemi SH, Earle J, Kanodia R, Kist D, Emamian ES, Patterson PH, Shi L, Sidwell R. Prenatal viral infection leads to pyramidal cell atrophy and macrocephaly in adulthood: implications for genesis of autism and schizophrenia. Cell Mol Neurobiol. 2002a;22:25–33. doi: 10.1023/a:1015337611258. [DOI] [PubMed] [Google Scholar]
  94. Fatemi SH, Emamian ES, Sidwell RW, Kist DA, Stary JM, Earle JA, Thuras P. Human influenza viral infection in utero alters glial fibrillary acidic protein immunoreactivity in the developing brains of neonatal mice. Mol Psychiatry. 2002b;7:633–640. doi: 10.1038/sj.mp.4001046. [DOI] [PubMed] [Google Scholar]
  95. Fatemi SH, Araghi-Niknam M, Laurence JA, Stary JM, Sidwell RW, Lee S. Glial fibrillary acidic protein and glutamic acid decarboxylase 65 and 67 kDa proteins are increased in brains of neonatal BALB/c mice following viral infection in utero. Schizophr Res. 2004;69:121–123. doi: 10.1016/S0920-9964(03)00175-0. [DOI] [PubMed] [Google Scholar]
  96. Fatemi SH, Pearce DA, Brooks AI, Sidwell RW. Prenatal viral infection in mouse causes differential expression of genes in brains of mouse progeny: a potential animal model for schizophrenia and autism. Synapse. 2005;57:91–99. doi: 10.1002/syn.20162. [DOI] [PubMed] [Google Scholar]
  97. Feldon J, Avnimelech-Gigus N, Weiner I. The effects of pre- and postweaning rearing conditions on latent inhibition and partial reinforcement extinction effect in male rats. Behav Neural Biol. 1990;53:189–204. doi: 10.1016/0163-1047(90)90407-w. [DOI] [PubMed] [Google Scholar]
  98. Fendt M, Lex A, Falkai P, Henn FA, Schmitt A. Behavioural alterations in rats following neonatal hypoxia and effects of clozapine: implications for schizophrenia. Pharmacopsychiatry. 2008;41:138–145. doi: 10.1055/s-2008-1058107. [DOI] [PubMed] [Google Scholar]
  99. Ferguson JN, Young LJ, Insel TR. The neuroendocrine basis of social recognition. Front Neuroendocrinol. 2002;23:200–224. doi: 10.1006/frne.2002.0229. [DOI] [PubMed] [Google Scholar]
  100. Feron F, Burne TH, Brown J, Smith E, McGrath JJ, Mackay-Sim A, Eyles DW. Developmental vitamin D3 deficiency alters the adult rat brain. Brain Res Bull. 2005;65:141–148. doi: 10.1016/j.brainresbull.2004.12.007. [DOI] [PubMed] [Google Scholar]
  101. Finamore TL, Port RL. Developmental stress disrupts habituation but spares prepulse inhibition in young rats. Physiol Behav. 2000;69:527–530. doi: 10.1016/s0031-9384(00)00205-5. [DOI] [PubMed] [Google Scholar]
  102. Finlay BL, Darlington RB. Linked regularities in the development and evolution of mammalian brains. Science. 1995;268:1578–1584. doi: 10.1126/science.7777856. [DOI] [PubMed] [Google Scholar]
  103. Flagstad P, Mork A, Glenthoj BY, van Beek J, Michael-Titus AT, Didriksen M. Disruption of neurogenesis on gestational day 17 in the rat causes behavioral changes relevant to positive and negative schizophrenia symptoms and alters amphetamine-induced dopamine release in nucleus accumbens. Neuropsychopharmacology. 2004;29:2052–2064. doi: 10.1038/sj.npp.1300516. [DOI] [PubMed] [Google Scholar]
  104. Flagstad P, Glenthoj BY, Didriksen M. Cognitive deficits caused by late gestational disruption of neurogenesis in rats: a preclinical model of schizophrenia. Neuropsychopharmacology. 2005;30:250–260. doi: 10.1038/sj.npp.1300625. [DOI] [PubMed] [Google Scholar]
  105. Flores G, Silva-Gomez AB, Ibanez O, Quirion R, Srivastava LK. Comparative behavioral changes in postpubertal rats after neonatal excitotoxic lesions of the ventral hippocampus and the prefrontal cortex. Synapse. 2005;56:147–153. doi: 10.1002/syn.20140. [DOI] [PubMed] [Google Scholar]
  106. Floresco SB, Zhang Y, Enomoto T. Neural circuits subserving behavioral flexibility and their relevance to schizophrenia. Behav Brain Res. 2009;204:396–409. doi: 10.1016/j.bbr.2008.12.001. [DOI] [PubMed] [Google Scholar]
  107. Fone KCF, Porkess MV. Behavioural and neurochemical effects of post-weaning social isolation in rodents – relevance to developmental neuropsychiatric disorders. Neurosci Biobehav Rev. 2008;32:1087–1102. doi: 10.1016/j.neubiorev.2008.03.003. [DOI] [PubMed] [Google Scholar]
  108. Fortier ME, Joober R, Luheshi GN, Boksa P. Maternal exposure to bacterial endotoxin during pregnancy enhances amphetamine-induced locomotion and startle responses in adult rat offspring. J Psychiatr Res. 2004;38:335–345. doi: 10.1016/j.jpsychires.2003.10.001. [DOI] [PubMed] [Google Scholar]
  109. Fortier ME, Luheshi GN, Boksa P. Effects of prenatal infection on prepulse inhibition in the rat depend on the nature of the infectious agent and the stage of pregnancy. Behav Brain Res. 2007;181:270–277. doi: 10.1016/j.bbr.2007.04.016. [DOI] [PubMed] [Google Scholar]
  110. Francois J, Ferrandon A, Koning E, Angst MJ, Sandner G, Nehlig A. Selective reorganization of GABAergic transmission in neonatal ventral hippocampal-lesioned rats. Int J Neuropsychopharmacol. 2009a;12:1097–1110. doi: 10.1017/S1461145709009985. [DOI] [PubMed] [Google Scholar]
  111. Francois J, Koning E, Ferrandon A, Sandner G, Nehlig A. Metabolic activity in the brain of juvenile and adult rats with a neonatal ventral hippocampal lesion. Hippocampus. 2009b doi: 10.1002/hipo.20686. (Epub ahead of print, July 31) [DOI] [PubMed] [Google Scholar]
  112. Frankle WG, Lerma J, Laruelle M. The synaptic hypothesis of schizophrenia. Neuron. 2003;39:205–216. doi: 10.1016/s0896-6273(03)00423-9. [DOI] [PubMed] [Google Scholar]
  113. Freedman R, Adler LE, Leonard S. Alternative phenotypes for the complex genetics of schizophrenia. Biol Psychiatry. 1999;45:551–558. doi: 10.1016/s0006-3223(98)00321-7. [DOI] [PubMed] [Google Scholar]
  114. Friston KJ, Frith CD. Schizophrenia: a disconnection syndrome? Clin Neurosci. 1995;3:89–97. [PubMed] [Google Scholar]
  115. Geyer MA, Markou A. Animal models of psychiatric disorders. In: Watson S, editor. Psychopharmacology: the fourth generation of progress CD-ROM version 3. Lippincott Williams & Wilkins; Philadelphia: 2002. [Google Scholar]
  116. Geyer MA, Moghaddam B. Animal models relevant to schizophrenia disorders. In: Charney D, Coyle J, Davis K, Nemeroff CB, editors. Neuropsychopharmacology: the fifth generation of progress. Lippincott Williams & Wilkins; New York: 2002. pp. 689–701. [Google Scholar]
  117. Geyer MA, Wilkinson LS, Humby T, Robbins TW. Isolation rearing of rats produces a deficit in prepulse inhibition of acoustic startle similar to that in schizophrenia. Biol Psychiatry. 1993;34:361–372. doi: 10.1016/0006-3223(93)90180-l. [DOI] [PubMed] [Google Scholar]
  118. Geyer MA, Krebs-Thomson K, Braff DL, Swerdlow NR. Pharmacological studies of prepulse inhibition models of sensorimotor gating deficits in schizophrenia: a decade in review. Psychopharmacology (Berl) 2001;156:117–154. doi: 10.1007/s002130100811. [DOI] [PubMed] [Google Scholar]
  119. Grecksch G, Rüthrich H, Höllt V, Becker A. Transient prenatal vitamin D deficiency is associated with changes of synaptic plasticity in the dentate gyrus in adult rats. Psychoneuroendocrinology. 2009;34S1:S258–S264. doi: 10.1016/j.psyneuen.2009.07.004. [DOI] [PubMed] [Google Scholar]
  120. Green MF, Nuechterlein KH, Gold JM, Barch DM, Cohen J, Essock S, Fenton WS, Frese F, Goldberg TE, Heaton RK, Keefe RS, Kern RS, Kraemer H, Stover E, Weinberger DR, Zalcman S, Marder SR. Approaching a consensus cognitive battery for clinical trials in schizophrenia: the NIMH-MATRICS conference to select cognitive domains and test criteria. Biol Psychiatry. 2004;56:301–307. doi: 10.1016/j.biopsych.2004.06.023. [DOI] [PubMed] [Google Scholar]
  121. Gue M, Bravard A, Meunier J, Veyrier R, Gaillet S, Recasens M, Maurice T. Sex differences in learning deficits induced by prenatal stress in juvenile rats. Behav Brain Res. 2004;150:149–157. doi: 10.1016/S0166-4328(03)00250-X. [DOI] [PubMed] [Google Scholar]
  122. Hagan JJ, Jones DN. Predicting drug efficacy for cognitive deficits in schizophrenia. Schizophr Bull. 2005;31:830–853. doi: 10.1093/schbul/sbi058. [DOI] [PubMed] [Google Scholar]
  123. Hall FS. Social deprivation of neonatal, adolescent, and adult rats has distinct neurochemical and behavioral consequences. Crit Rev Neurobiol. 1998;12:129–162. doi: 10.1615/critrevneurobiol.v12.i1-2.50. [DOI] [PubMed] [Google Scholar]
  124. Hall FS, Humby T, Wilkinson LS, Robbins TW. The effects of isolation-rearing on preference by rats for a novel environment. Physiol Behav. 1997;62:299–303. doi: 10.1016/s0031-9384(97)00117-0. [DOI] [PubMed] [Google Scholar]
  125. Hall FS, Wilkinson LS, Humby T, Inglis W, Kendall DA, Marsden CA, Robbins TW. Isolation rearing in rats: pre- and postsynaptic changes in striatal dopaminergic systems. Pharmacol Biochem Behav. 1998;59:859–872. doi: 10.1016/s0091-3057(97)00510-8. [DOI] [PubMed] [Google Scholar]
  126. Harms LR, Eyles DW, McGrath JJ, Mackay-Sim A, Burne TH. Developmental vitamin D deficiency alters adult behaviour in 129/SvJ and C57BL/6J mice. Behav Brain Res. 2008;187:343–350. doi: 10.1016/j.bbr.2007.09.032. [DOI] [PubMed] [Google Scholar]
  127. Harrison PJ, Weinberger DR. Schizophrenia genes, gene expression, and neuropathology: on the matter of their convergence. Mol Psychiatry. 2005;10:40–68. doi: 10.1038/sj.mp.4001558. image 5. [DOI] [PubMed] [Google Scholar]
  128. Harte MK, Powell SB, Swerdlow NR, Geyer MA, Reynolds GP. Deficits in parvalbumin and calbindin immunoreactive cells in the hippocampus of isolation reared rats. J Neural Transm. 2007;114:893–898. doi: 10.1007/s00702-007-0627-6. [DOI] [PubMed] [Google Scholar]
  129. Holmes A, le Guisquet AM, Vogel E, Millstein RA, Leman S, Belzung C. Early life genetic, epigenetic and environmental factors shaping emotionality in rodents. Neurosci Biobehav Rev. 2005;29:1335–1346. doi: 10.1016/j.neubiorev.2005.04.012. [DOI] [PubMed] [Google Scholar]
  130. Holtze M, Asp L, Schwieler L, Engberg G, Karlsson H. Induction of the kynurenine pathway by neurotropic influenza A virus infection. J Neurosci Res. 2008;86:3674–3683. doi: 10.1002/jnr.21799. [DOI] [PubMed] [Google Scholar]
  131. Huang Z, Liu J, Cheung P-Y, Chen C. Long-term cognitive impairment and myelination deficiency in a rat model of perinatal hypoxic-ischemic brain injury. Brain Res. 2009;1301:100–109. doi: 10.1016/j.brainres.2009.09.006. [DOI] [PubMed] [Google Scholar]
  132. Hultman CM, Ohman A, Cnattingius S, Wieselgren IM, Lindstrom LH. Prenatal and neonatal risk factors for schizophrenia. Br J Psychiatry. 1997;170:128–133. doi: 10.1192/bjp.170.2.128. [DOI] [PubMed] [Google Scholar]
  133. Hunt PS. Neonatal treatment with a competitive NMDA antagonist results in response-specific disruption of conditioned fear in preweanling rats. Psychopharmacology (Berl) 2006;185:179–187. doi: 10.1007/s00213-005-0291-1. [DOI] [PubMed] [Google Scholar]
  134. Huot RL, Plotsky PM, Lenox RH, McNamara RK. Neonatal maternal separation reduces hippocampal mossy fiber density in adult Long Evans rats. Brain Res. 2002;950:52–63. doi: 10.1016/s0006-8993(02)02985-2. [DOI] [PubMed] [Google Scholar]
  135. Ibi D, Takuma K, Koike H, Mizoguchi H, Tsuritani K, Kuwahara Y, Kamei H, Nagai T, Yoneda Y, Nabeshima T, Yamada K. Social isolation rearing-induced impairment of the hippocampal neurogenesis is associated with deficits in spatial memory and emotion-related behaviors in juvenile mice. J Neurochem. 2008;105:921–932. doi: 10.1111/j.1471-4159.2007.05207.x. [DOI] [PubMed] [Google Scholar]
  136. Ibi D, Nagai T, Kitahara Y, Mizoguchi H, Koike H, Shiraki A, Takuma K, Kamei H, Noda Y, Nitta A, Nabeshima T, Yoneda Y, Yamada K. Neonatal polyI:C treatment in mice results in schizophrenia-like behavioral and neurochemical abnormalities in adulthood. Neurosci Res. 2009;64:297–305. doi: 10.1016/j.neures.2009.03.015. [DOI] [PubMed] [Google Scholar]
  137. Ibi D, Nagai T, Koike H, Kitahara Y, Mizoguchi H, Niwa M, Jaaro-Peled H, Nitta A, Yoneda Y, Nabeshima T, Sawa A, Yamada K. Combined effect of neonatal immune activation and mutant DISC1 on phenotypic changes in adulthood. Behav Brain Res. 2010;206:32–37. doi: 10.1016/j.bbr.2009.08.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Jakob H, Beckmann H. Prenatal developmental disturbances in the limbic allocortex in schizophrenics. J Neural Transm. 1986;65:303–326. doi: 10.1007/BF01249090. [DOI] [PubMed] [Google Scholar]
  139. Jenkins TA, Harte MK, Stenson G, Reynolds GP. Neonatal lipopolysaccharide induces pathological changes in parvalbumin immunoreactivity in the hippocampus of the rat. Behav Brain Res. 2009;205:355–359. doi: 10.1016/j.bbr.2009.07.014. [DOI] [PubMed] [Google Scholar]
  140. Johnstone EC, Ebmeier KP, Miller P, Owens DG, Lawrie SM. Predicting schizophrenia: findings from the Edinburgh High-Risk Study. Br J Psychiatry. 2005;186:18–25. doi: 10.1192/bjp.186.1.18. [DOI] [PubMed] [Google Scholar]
  141. Jones GH, Robbins TW, Marsden CA. Isolation-rearing retards the acquisition of schedule-induced polydipsia in rats. Physiol Behav. 1989;45:71–77. doi: 10.1016/0031-9384(89)90167-4. [DOI] [PubMed] [Google Scholar]
  142. Jones GH, Marsden CA, Robbins TW. Increased sensitivity to amphetamine and reward-related stimuli following social isolation in rats: possible disruption of dopamine-dependent mechanisms of the nucleus accumbens. Psychopharmacology (Berl) 1990;102:364–372. doi: 10.1007/BF02244105. [DOI] [PubMed] [Google Scholar]
  143. Jones GH, Hernandez TD, Kendall DA, Marsden CA, Robbins TW. Dopaminergic and serotonergic function following isolation rearing in rats: study of behavioural responses and postmortem and in vivo neurochemistry. Pharmacol Biochem Behav. 1992;43:17–35. doi: 10.1016/0091-3057(92)90635-s. [DOI] [PubMed] [Google Scholar]
  144. Jones DNC, Gartlon JE, Minassian A, Perry W, Geyer MA. Developing new drugs for schizophrenia: from animals to the clinic. In: McArthur R, Borsini R, editors. Animal and translational models for CNS drug discovery: psychiatric disorders. Elsevier; New York: 2008. pp. 199–262. [Google Scholar]
  145. Juarez I, De La Cruz F, Zamudio S, Flores G. Cesarean plus anoxia at birth induces hyperresponsiveness to locomotor activity by dopamine D2 agonist. Synapse. 2005;58:236–242. doi: 10.1002/syn.20203. [DOI] [PubMed] [Google Scholar]
  146. Kapoor A, Kostaki A, Janus C, Matthews SG. The effects of prenatal stress on learning in adult offspring is dependent on the timing of the stressor. Behav Brain Res. 2009;197:144–149. doi: 10.1016/j.bbr.2008.08.018. [DOI] [PubMed] [Google Scholar]
  147. Kendell RE, Adams W. Exposure to sunlight, vitamin D and schizophrenia. Schizophr Res. 2002;54:193–198. doi: 10.1016/s0920-9964(01)00264-x. [DOI] [PubMed] [Google Scholar]
  148. Kesby JP, Burne TH, McGrath JJ, Eyles DW. Developmental vitamin D deficiency alters MK 801-induced hyperlocomotion in the adult rat: an animal model of schizophrenia. Biol Psychiatry. 2006;60:591–596. doi: 10.1016/j.biopsych.2006.02.033. [DOI] [PubMed] [Google Scholar]
  149. Keshavan MS, Hogarty GE. Brain maturational processes and delayed onset in schizophrenia. Dev Psychopathol. 1999;11:525–543. doi: 10.1017/s0954579499002199. [DOI] [PubMed] [Google Scholar]
  150. 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. doi: 10.1001/archgenpsychiatry.2007.20. [DOI] [PubMed] [Google Scholar]
  151. Kietzman M, Spring B, Zubin J. Perception, cognition, and information processing. In: Kaplan H, Sadock B, editors. Comprehensive textbook of psychiatry IV. Williams & Williams; Baltimore: 1985. [Google Scholar]
  152. Koenig JI. Schizophrenia: a unique translational opportunity in behavioral neuroendocrinology. Horm Behav. 2006;50:602–611. doi: 10.1016/j.yhbeh.2006.06.027. [DOI] [PubMed] [Google Scholar]
  153. Koenig JI, Kirkpatrick B, Lee P. Glucocorticoid hormones and early brain development in schizophrenia. Neuropsychopharmacology. 2002;27:309–318. doi: 10.1016/S0893-133X(01)00396-7. [DOI] [PubMed] [Google Scholar]
  154. Koenig JI, Elmer GI, Shepard PD, Lee PR, Mayo C, Joy B, Hercher E, Brady DL. Prenatal exposure to a repeated variable stress paradigm elicits behavioral and neuroendocrinological changes in the adult offspring: potential relevance to schizophrenia. Behav Brain Res. 2005;156:251–261. doi: 10.1016/j.bbr.2004.05.030. [DOI] [PubMed] [Google Scholar]
  155. Krech D, Rosenzweig MR, Bennett EL. Relations between chemistry and problem-solving among rats raised in enriched and impoverished environments. J Comp Physiol Psychol. 1962;55:801–807. doi: 10.1037/h0044220. [DOI] [PubMed] [Google Scholar]
  156. Lapiz MD, Mateo Y, Parker T, Marsden C. Effects of noradrenaline depletion in the brain on response on novelty in isolation-reared rats. Psychopharmacology (Berl) 2000;152:312–320. doi: 10.1007/s002130000534. [DOI] [PubMed] [Google Scholar]
  157. Laruelle M, Abi-Dargham A, van Dyck CH, Gil R, D’Souza CD, Erdos J, McCance E, Rosenblatt W, Fingado C, Zoghbi SS, Baldwin RM, Seibyl JP, Krystal JH, Charney DS, Innis RB. Single photon emission computerized tomography imaging of amphetamine-induced dopamine release in drug-free schizophrenic subjects. Proc Natl Acad Sci USA. 1996;93:9235–9240. doi: 10.1073/pnas.93.17.9235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Le Pen G, Moreau JL. Disruption of prepulse inhibition of startle reflex in a neurodevelopmental model of schizophrenia: reversal by clozapine, olanzapine and risperidone but not by haloperidol. Neuropsychopharmacology. 2002;27:1–11. doi: 10.1016/S0893-133X(01)00383-9. [DOI] [PubMed] [Google Scholar]
  159. Le Pen G, Grottick AJ, Higgins GA, Martin JR, Jenck F, Moreau JL. Spatial and associative learning deficits induced by neonatal excitotoxic hippocampal damage in rats: further evaluation of an animal model of schizophrenia. Behav Pharmacol. 2000;11:257–268. doi: 10.1097/00008877-200006000-00009. [DOI] [PubMed] [Google Scholar]
  160. Le Pen G, Kew J, Alberati D, Borroni E, Heitz MP, Moreau JL. Prepulse inhibition deficits of the startle reflex in neonatal ventral hippocampal-lesioned rats: reversal by glycine and a glycine transporter inhibitor. Biol Psychiatry. 2003;54:1162–1170. doi: 10.1016/s0006-3223(03)00374-3. [DOI] [PubMed] [Google Scholar]
  161. Le Pen G, Gourevitch R, Hazane F, Hoareau C, Jay TM, Krebs MO. Peri-pubertal maturation after developmental disturbance: a model for psychosis onset in the rat. Neuroscience. 2006;143:395–405. doi: 10.1016/j.neuroscience.2006.08.004. [DOI] [PubMed] [Google Scholar]
  162. Lee PR, Brady DL, Shapiro RA, Dorsa DM, Koenig JI. Prenatal stress generates deficits in rat social behavior: reversal by oxytocin. Brain Res. 2007;1156:152–167. doi: 10.1016/j.brainres.2007.04.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Lehmann J, Stohr T, Feldon J. Long-term effects of prenatal stress experiences and postnatal maternal separation on emotionality and attentional processes. Behav Brain Res. 2000;107:133–144. doi: 10.1016/s0166-4328(99)00122-9. [DOI] [PubMed] [Google Scholar]
  164. Lei G, Anastasio NC, Fu Y, Neugebauer V, Johnson KM. Activation of dopamine D1 receptors blocks phencyclidine-induced neurotoxicity by enhancing N-methyl-D-aspartate receptor-mediated synaptic strength. J Neurochem. 2009;109:1017–1030. doi: 10.1111/j.1471-4159.2009.06030.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Lemaire V, Koehl M, Le Moal M, Abrous DN. Prenatal stress produces learning deficits associated with an inhibition of neurogenesis in the hippocampus. Proc Natl Acad Sci USA. 2000;97:11032–11037. doi: 10.1073/pnas.97.20.11032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Lena I, Chessel A, Le Pen G, Krebs MO, Garcia R. Alterations in prefrontal glutamatergic and noradrenergic systems following MK-801 administration in rats prenatally exposed to methylazoxymethanol at gestational day 17. Psychopharmacology (Berl) 2007;192:373–383. doi: 10.1007/s00213-007-0719-x. [DOI] [PubMed] [Google Scholar]
  167. Lewis DA, Levitt P. Schizophrenia as a disorder of neurodevelopment. Ann Rev Neurosci. 2002;25:409–432. doi: 10.1146/annurev.neuro.25.112701.142754. [DOI] [PubMed] [Google Scholar]
  168. Li Q, Cheung C, Wei R, Hui ES, Feldon J, Meyer U, Chung S, Chua SE, Sham PC, Wu EX, McAlonan GM. Prenatal immune challenge is an environmental risk factor for brain and behavior change relevant to schizophrenia: evidence from MRI in a mouse model. PLoS One. 2009;4:e6354. doi: 10.1371/journal.pone.0006354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Licinio J, Seibyl JP, Altemus M, Charney DS, Krystal JH. Elevated CSF levels of interleukin-2 in neuroleptic-free schizophrenic patients. Am J Psychiatry. 1993;150:1408–1410. doi: 10.1176/ajp.150.9.1408. [DOI] [PubMed] [Google Scholar]
  170. Lipska BK. Using animal models to test a neurodevelopmental hypothesis of schizophrenia. J Psychiatry Neurosci. 2004;29:282–286. [PMC free article] [PubMed] [Google Scholar]
  171. Lipska BK, Weinberger DR. Delayed effects of neonatal hippocampal damage on haloperidol-induced catalepsy and apomorphine-induced stereotypic behaviors in the rat. Brain Res Dev Brain Res. 1993;75:213–222. doi: 10.1016/0165-3806(93)90026-7. [DOI] [PubMed] [Google Scholar]
  172. Lipska BK, Weinberger DR. Gonadectomy does not prevent novelty or drug-induced motor hyperresponsiveness in rats with neonatal hippocampal damage. Brain Res Dev Brain Res. 1994a;78:253–258. doi: 10.1016/0165-3806(94)90033-7. [DOI] [PubMed] [Google Scholar]
  173. Lipska BK, Weinberger DR. Subchronic treatment with haloperidol and clozapine in rats with neonatal excitotoxic hippocampal damage. Neuropsychopharmacology. 1994b;10:199–205. doi: 10.1038/npp.1994.22. [DOI] [PubMed] [Google Scholar]
  174. Lipska BK, Weinberger DR. To model a psychiatric disorder in animals: schizophrenia as a reality test. Neuropsychopharmacology. 2000;23:223–239. doi: 10.1016/S0893-133X(00)00137-8. [DOI] [PubMed] [Google Scholar]
  175. Lipska BK, Jaskiw GE, Weinberger DR. Postpubertal emergence of hyperresponsiveness to stress and to amphetamine after neonatal excitotoxic hippocampal damage: a potential animal model of schizophrenia. Neuropsychopharmacology. 1993;9:67–75. doi: 10.1038/npp.1993.44. [DOI] [PubMed] [Google Scholar]
  176. Lipska BK, Swerdlow NR, Geyer MA, Jaskiw GE, Braff DL, Weinberger DR. Neonatal excitotoxic hippocampal damage in rats causes post-pubertal changes in prepulse inhibition of startle and its disruption by apomorphine. Psychopharmacology (Berl) 1995;122:35–43. doi: 10.1007/BF02246439. [DOI] [PubMed] [Google Scholar]
  177. Lipska BK, Aultman JM, Verma A, Weinberger DR, Moghaddam B. Neonatal damage of the ventral hippocampus impairs working memory in the rat. Neuropsychopharmacology. 2002a;27:47–54. doi: 10.1016/S0893-133X(02)00282-8. [DOI] [PubMed] [Google Scholar]
  178. Lipska BK, Halim ND, Segal PN, Weinberger DR. Effects of reversible inactivation of the neonatal ventral hippocampus on behavior in the adult rat. J Neurosci. 2002b;22:2835–2842. doi: 10.1523/JNEUROSCI.22-07-02835.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Lipska BK, Lerman DN, Khaing ZZ, Weickert CS, Weinberger DR. Gene expression in dopamine and GABA systems in an animal model of schizophrenia: effects of antipsychotic drugs. Eur J Neurosci. 2003;18:391–402. doi: 10.1046/j.1460-9568.2003.02738.x. [DOI] [PubMed] [Google Scholar]
  180. Lodge DJ, Grace AA. Aberrant hippocampal activity underlies the dopamine dysregulation in an animal model of schizophrenia. J Neurosci. 2007;27:11424–11430. doi: 10.1523/JNEUROSCI.2847-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Lodge DJ, Grace AA. Gestational methylazoxymethanol acetate administration: a developmental disruption model of schizophrenia. Behav Brain Res. 2009;204:306–312. doi: 10.1016/j.bbr.2009.01.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Lodge DJ, Behrens MM, Grace AA. A loss of parvalbumin-containing interneurons is associated with diminished oscillatory activity in an animal model of schizophrenia. J Neurosci. 2009;29:2344–2354. doi: 10.1523/JNEUROSCI.5419-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Machon RA, Mednick SA, Schulsinger F. The interaction of seasonality, place of birth, genetic rish and subsequent schizophrenia in a high risk sample. Br J Psychiatry. 1983;143:383–388. doi: 10.1192/bjp.143.4.383. [DOI] [PubMed] [Google Scholar]
  184. Mallard EC, Rehn A, Rees S, Tolcos M, Copolov D. Ventriculomegaly and reduced hippocampal volume following intrauterine growth-restriction: implications for the aetiology of schizophrenia. Schizophr Res. 1999;40:11–21. doi: 10.1016/s0920-9964(99)00041-9. [DOI] [PubMed] [Google Scholar]
  185. Mallard C, Loeliger M, Copolov D, Rees S. Reduced number of neurons in the hippocampus and the cerebellum in the postnatal guinea-pig following intrauterine growth-restriction. Neuroscience. 2000;100:327–333. doi: 10.1016/s0306-4522(00)00271-2. [DOI] [PubMed] [Google Scholar]
  186. Marcelis M, Navarro-Mateu F, Murray R, Selten JP, Van Os J. Urbanization and psychosis: a study of 1942–1978 birth cohorts in The Netherlands. Psychol Med. 1998;28:871–879. doi: 10.1017/s0033291798006898. [DOI] [PubMed] [Google Scholar]
  187. Marquis JP, Goulet S, Doré FY. Neonatal lesions of the ventral hippocampus in rats lead to prefrontal cognitive deficits at two maturational stages. Neuroscience. 2006;140:759–767. doi: 10.1016/j.neuroscience.2006.02.048. [DOI] [PubMed] [Google Scholar]
  188. Marquis JP, Goulet S, Dore FY. Neonatal ventral hippocampus lesions disrupt extra-dimensional shift and alter dendritic spine density in the medial prefrontal cortex of juvenile rats. Neurobiol Learn Mem. 2008;90:339–346. doi: 10.1016/j.nlm.2008.04.005. [DOI] [PubMed] [Google Scholar]
  189. McClellan J, Breiger D, McCurry C, Hlastala SA. Premorbid functioning in early-onset psychotic disorders. J Am Acad Child Adolesc Psychiatry. 2003;42:666–672. doi: 10.1097/01.CHI.0000046844.56865.6B. [DOI] [PubMed] [Google Scholar]
  190. McGlashan TH, Hoffman RE. Schizophrenia as a disorder of developmentally reduced synaptic connectivity. Arch Gen Psychiatry. 2000;57:637–648. doi: 10.1001/archpsyc.57.7.637. [DOI] [PubMed] [Google Scholar]
  191. McGrath J. Hypothesis: is low prenatal vitamin D a risk-modifying factor for schizophrenia? Schizophr Res. 1999;40:173–177. doi: 10.1016/s0920-9964(99)00052-3. [DOI] [PubMed] [Google Scholar]
  192. McGrath J, Eyles D, Mowry B, Yolken R, Buka S. Low maternal vitamin D as a risk factor for schizophrenia: a pilot study using banked sera. Schizophr Res. 2003;63:73–78. doi: 10.1016/s0920-9964(02)00435-8. [DOI] [PubMed] [Google Scholar]
  193. McGrath J, Saari K, Hakko H, Jokelainen J, Jones P, Jarvelin MR, Chant D, Isohanni M. Vitamin D supplementation during the first year of life and risk of schizophrenia: a Finnish birth cohort study. Schizophr Res. 2004;67:237–245. doi: 10.1016/j.schres.2003.08.005. [DOI] [PubMed] [Google Scholar]
  194. McGrath J, Iwazaki T, Eyles D, Burne T, Cui X, Ko P, Matsumoto I. Protein expression in the nucleus accumbens of rats exposed to developmental vitamin D deficiency. PLoS One. 2008;3:e2383. doi: 10.1371/journal.pone.0002383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. McLean S, Grayson B, Harris M, Protheroe C, Bate S, Woolley M, Neill J. Isolation rearing impairs novel object recognition and attentional set-shifting performance in female rats. J Psychopharmacol. 2010;24:57–63. doi: 10.1177/0269881108093842. [DOI] [PubMed] [Google Scholar]
  196. Meaney MJ, Mitchell JB, Aitken DH, Bhatnagar S, Bodnoff SR, Iny LJ, Sarrieau A. The effects of neonatal handling on the development of the adrenocortical response to stress: implications for neuropathology and cognitive deficits in later life. Psychoneuroendocrinology. 1991;16:85–103. doi: 10.1016/0306-4530(91)90072-2. [DOI] [PubMed] [Google Scholar]
  197. Meaney MJ, Bhatnagar S, Larocque S, McCormick C, Shanks N, Sharma S, Smythe J, Viau V, Plotsky PM. Individual differences in the hypothalamic-pituitary-adrenal stress response and the hypothalamic CRF system. Ann N Y Acad Sci. 1993;697:70–85. doi: 10.1111/j.1749-6632.1993.tb49924.x. [DOI] [PubMed] [Google Scholar]
  198. Mednick SA, Machon RA, Huttunen MO, Bonett D. Adult schizophrenia following prenatal exposure to an influenza epidemic. Arch Gen Psychiatry. 1988;45:189–192. doi: 10.1001/archpsyc.1988.01800260109013. [DOI] [PubMed] [Google Scholar]
  199. Meyer U, Feldon J. Epidemiology-driven neurodevelopmental animal models of schizophrenia. Prog Neurobiol. 2009a doi: 10.1016/j.pneurobio.2009.10.018. (Epub ahead of print, Oct 24) [DOI] [PubMed] [Google Scholar]
  200. Meyer U, Feldon J. Neural basis of psychosis-related behaviour in the infection model of schizophrenia. Behav Brain Res. 2009b;204:322–334. doi: 10.1016/j.bbr.2008.12.022. [DOI] [PubMed] [Google Scholar]
  201. Meyer U, Feldon J, Schedlowski M, Yee BK. Towards an immuno-precipitated neurodevelopmental animal model of schizophrenia. Neurosci Biobehav Rev. 2005;29:913–947. doi: 10.1016/j.neubiorev.2004.10.012. [DOI] [PubMed] [Google Scholar]
  202. Meyer U, Feldon J, Schedlowski M, Yee BK. Immunological stress at the maternal–foetal interface: a link between neurodevelopment and adult psychopathology. Brain Behav Immun. 2006a;20:378–388. doi: 10.1016/j.bbi.2005.11.003. [DOI] [PubMed] [Google Scholar]
  203. Meyer U, Nyffeler M, Engler A, Urwyler A, Schedlowski M, Knuesel I, Yee BK, Feldon J. The time of prenatal immune challenge determines the specificity of inflammation-mediated brain and behavioral pathology. J Neurosci. 2006b;26:4752–4762. doi: 10.1523/JNEUROSCI.0099-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  204. Meyer U, Schwendener S, Feldon J, Yee BK. Prenatal and postnatal maternal contributions in the infection model of schizophrenia. Exp Brain Res. 2006c;173:243–257. doi: 10.1007/s00221-006-0419-5. [DOI] [PubMed] [Google Scholar]
  205. Meyer U, Murray PJ, Urwyler A, Yee BK, Schedlowski M, Feldon J. Adult behavioral and pharmacological dysfunctions following disruption of the fetal brain balance between pro-inflammatory and IL-10-mediated anti-inflammatory signaling. Mol Psychiatry. 2008a;13:208–221. doi: 10.1038/sj.mp.4002042. [DOI] [PubMed] [Google Scholar]
  206. Meyer U, Nyffeler M, Schwendener S, Knuesel I, Yee BK, Feldon J. Relative prenatal and postnatal maternal contributions to schizophrenia-related neurochemical dysfunction after in utero immune challenge. Neuropsychopharmacology. 2008b;33:441–456. doi: 10.1038/sj.npp.1301413. [DOI] [PubMed] [Google Scholar]
  207. Meyer U, Nyffeler M, Yee BK, Knuesel I, Feldon J. Adult brain and behavioral pathological markers of prenatal immune challenge during early/middle and late fetal development in mice. Brain Behav Immun. 2008c;22:469–486. doi: 10.1016/j.bbi.2007.09.012. [DOI] [PubMed] [Google Scholar]
  208. Meyer U, Spoerri E, Yee BK, Schwarz MJ, Feldon J. Evaluating early preventive antipsychotic and antidepressant drug treatment in an infection-based neurodevelopmental mouse model of schizophrenia. Schizophr Bull. 2008d doi: 10.1093/schbul/sbn131. (Epub ahead of print, Oct 8) [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Meyer U, Feldon J, Fatemi SH. In-vivo rodent models for the experimental investigation of prenatal immune activation effects in neurodevelopmental brain disorders. Neurosci Biobehav Rev. 2009;33:1061–1079. doi: 10.1016/j.neubiorev.2009.05.001. [DOI] [PubMed] [Google Scholar]
  210. Miller P, Byrne M, Hodges A, Lawrie SM, Owens DG, Johnstone EC. Schizotypal components in people at high risk of developing schizophrenia: early findings from the Edinburgh high-risk study. Br J Psychiatry. 2002;180:179–184. doi: 10.1192/bjp.180.2.179. [DOI] [PubMed] [Google Scholar]
  211. Millstein RA, Ralph RJ, Yang RJ, Holmes A. Effects of repeated maternal separation on prepulse inhibition of startle across inbred mouse strains. Genes Brain Behav. 2006;5:346–354. doi: 10.1111/j.1601-183X.2005.00172.x. [DOI] [PubMed] [Google Scholar]
  212. Mino Y, Oshima I. Seasonality of birth in patients with schizophrenia in Japan. Psychiatry Clin Neurosci. 2006;60:249–252. doi: 10.1111/j.1440-1819.2006.01493.x. [DOI] [PubMed] [Google Scholar]
  213. Mirnics K, Middleton FA, Lewis DA, Levitt P. Analysis of complex brain disorders with gene expression microarrays: schizophrenia as a disease of the synapse. Trends Neurosci. 2001;24:479–486. doi: 10.1016/s0166-2236(00)01862-2. [DOI] [PubMed] [Google Scholar]
  214. Mittleman BB, Castellanos FX, Jacobsen LK, Rapoport JL, Swedo SE, Shearer GM. Cerebrospinal fluid cytokines in pediatric neuropsychiatric disease. J Immunol. 1997;159:2994–2999. [PubMed] [Google Scholar]
  215. Moore H, Jentsch JD, Ghajarnia M, Geyer MA, Grace AA. A neurobehavioral systems analysis of adult rats exposed to methylazoxymethanol acetate on E17: implications for the neuropathology of schizophrenia. Biol Psychiatry. 2006;60:253–264. doi: 10.1016/j.biopsych.2006.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Mouri A, Noda Y, Enomoto T, Nabeshima T. Phencyclidine animal models of schizophrenia: approaches from abnormality of glutamatergic neurotransmission and neurodevelopment. Neurochem Int. 2007;51:173–184. doi: 10.1016/j.neuint.2007.06.019. [DOI] [PubMed] [Google Scholar]
  217. Moy SS, Nadler JJ, Perez A, Barbaro RP, Johns JM, Magnuson TR, Piven J, Crawley JN. Sociability and preference for social novelty in five inbred strains: an approach to assess autistic-like behavior in mice. Genes Brain Behav. 2004;3:287–302. doi: 10.1111/j.1601-1848.2004.00076.x. [DOI] [PubMed] [Google Scholar]
  218. Murray RM, Lewis SW. Is schizophrenia a neurodevelopmental disorder? Br Med J (Clin Res Ed) 1988;296:263. doi: 10.1136/bmj.296.6614.63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. Nadri C, Belmaker RH, Agam G. Oxygen restriction of neonate rats elevates neuregulin-1alpha isoform levels: possible relationship to schizophrenia. Neurochem Int. 2007;51:447–450. doi: 10.1016/j.neuint.2007.03.013. [DOI] [PubMed] [Google Scholar]
  220. Nakatani-Pawlak A, Yamaguchi K, Tatsumi Y, Mizoguchi H, Yoneda Y. Neonatal phencyclidine treatment in mice induces behavioral, histological and neurochemical abnormalities in adulthood. Biol Pharm Bull. 2009;32:1576–1583. doi: 10.1248/bpb.32.1576. [DOI] [PubMed] [Google Scholar]
  221. Nawa H, Takahashi M, Patterson PH. Cytokine and growth factor involvement in schizophrenia – support for the developmental model. Mol Psychiatry. 2000;5:594–603. doi: 10.1038/sj.mp.4000730. [DOI] [PubMed] [Google Scholar]
  222. Nestor PG, O’Donnell BF. The mind adrift: attentional dysregulation in schizophrenia. In: Parasuraman R, editor. The attentive brain. MIT Press; Cambridge, MA: 1998. pp. 527–546. [Google Scholar]
  223. Nolan Y, Vereker E, Lynch AM, Lynch MA. Evidence that lipopolysaccharide-induced cell death is mediated by accumulation of reactive oxygen species and activation of p38 in rat cortex and hippocampus. Exp Neurol. 2003;184:794–804. doi: 10.1016/S0014-4886(03)00301-7. [DOI] [PubMed] [Google Scholar]
  224. Nudmamud S, Reynolds LM, Reynolds GP. N-acetylaspartate and N-Acetylaspartylglutamate deficits in superior temporal cortex in schizophrenia and bipolar disorder: a postmortem study. Biol Psychiatry. 2003;53:1138–1141. doi: 10.1016/s0006-3223(02)01742-0. [DOI] [PubMed] [Google Scholar]
  225. Nuechterlein KH, Barch DM, Gold JM, Goldberg TE, Green MF, Heaton RK. Identification of separable cognitive factors in schizophrenia. Schizophr Res. 2004;72:29–39. doi: 10.1016/j.schres.2004.09.007. [DOI] [PubMed] [Google Scholar]
  226. Nyffeler M, Meyer U, Yee BK, Feldon J, Knuesel I. Maternal immune activation during pregnancy increases limbic GABAA receptor immunoreactivity in the adult offspring: implications for schizophrenia. Neuroscience. 2006;143:51–62. doi: 10.1016/j.neuroscience.2006.07.029. [DOI] [PubMed] [Google Scholar]
  227. O’Callaghan E, Sham P, Takei N, Glover G, Murray RM. Schizophrenia after prenatal exposure to 1957 A2 influenza epidemic. Lancet. 1991;337:1248–1250. doi: 10.1016/0140-6736(91)92919-s. [DOI] [PubMed] [Google Scholar]
  228. O’Donnell P, Lewis BL, Weinberger DR, Lipska BK. Neonatal hippocampal damage alters electrophysiological properties of prefrontal cortical neurons in adult rats. Cereb Cortex. 2002;12:975–982. doi: 10.1093/cercor/12.9.975. [DOI] [PubMed] [Google Scholar]
  229. O’Loan J, Eyles DW, Kesby J, Ko P, McGrath JJ, Burne TH. Vitamin D deficiency during various stages of pregnancy in the rat; its impact on development and behaviour in adult offspring. Psychoneuroendocrinology. 2007;32:227–234. doi: 10.1016/j.psyneuen.2006.12.006. [DOI] [PubMed] [Google Scholar]
  230. Orzack MH, Kornetsky C. Attention dysfunction in chronic schizophrenia. Arch Gen Psychiatry. 1966;14:323–326. doi: 10.1001/archpsyc.1966.01730090099015. [DOI] [PubMed] [Google Scholar]
  231. Owen MJ, Lewis SW. Risk factors in schizophrenia. Br J Psychiatry. 1988;153:407. doi: 10.1192/bjp.153.3.407a. [DOI] [PubMed] [Google Scholar]
  232. Ozawa K, Hashimoto K, Kishimoto T, Shimizu E, Ishikura H, Iyo M. Immune activation during pregnancy in mice leads to dopaminergic hyperfunction and cognitive impairment in the offspring: a neurodevelopmental animal model of schizophrenia. Biol Psychiatry. 2006;59:546–554. doi: 10.1016/j.biopsych.2005.07.031. [DOI] [PubMed] [Google Scholar]
  233. Özer S, Ulusahin A, Ulusoy S, Okur H, Coskun T, TuncalI T, Gögüs A, Akarsu AN. Is vitamin D hypothesis for schizophrenia valid? Independent segregation of psychosis in a family with vitamin-D-dependent rickets type IIA. Prog Neuropsychopharmacol Biol Psychiatry. 2004;28:255–266. doi: 10.1016/j.pnpbp.2003.10.002. [DOI] [PubMed] [Google Scholar]
  234. Palmer AA, Printz DJ, Butler PD, Dulawa SC, Printz MP. Prenatal protein deprivation in rats induces changes in prepulse inhibition and NMDA receptor binding. Brain Res. 2004;996:193–201. doi: 10.1016/j.brainres.2003.09.077. [DOI] [PubMed] [Google Scholar]
  235. Palmer AA, Brown AS, Keegan D, Siska LD, Susser E, Rotrosen J, Butler PD. Prenatal protein deprivation alters dopamine-mediated behaviors and dopaminergic and glutamatergic receptor binding. Brain Res. 2008;1237:62–74. doi: 10.1016/j.brainres.2008.07.089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Pascual R, Zamora-Leon SP, Valero-Cabre A. Effects of postweaning social isolation and re-socialization on the expression of vasoactive intestinal peptide (VIP) and dendritic development in the medial prefrontal cortex of the rat. Acta Neurobiol Exp (Wars) 2006;66:7–14. doi: 10.55782/ane-2006-1582. [DOI] [PubMed] [Google Scholar]
  237. Patterson PH. Immune involvement in schizophrenia and autism: etiology, pathology and animal models. Behav Brain Res. 2009;204:313–321. doi: 10.1016/j.bbr.2008.12.016. [DOI] [PubMed] [Google Scholar]
  238. Paulus MP, Bakshi VP, Geyer MA. Isolation rearing affects sequential organization of motor behavior in post-pubertal but not pre-pubertal Lister and Sprague-Dawley rats. Behav Brain Res. 1998;94:271–280. doi: 10.1016/s0166-4328(97)00158-7. [DOI] [PubMed] [Google Scholar]
  239. Penschuck S, Flagstad P, Didriksen M, Leist M, Michael-Titus AT. Decrease in parvalbumin-expressing neurons in the hippocampus and increased phencyclidine-induced locomotor activity in the rat methylazoxymethanol (MAM) model of schizophrenia. Eur J Neurosci. 2006;23:279–284. doi: 10.1111/j.1460-9568.2005.04536.x. [DOI] [PubMed] [Google Scholar]
  240. Pereira LO, Arteni NS, Petersen RC, da Rocha AP, Achaval M, Netto CA. Effects of daily environmental enrichment on memory deficits and brain injury following neonatal hypoxia-ischemia in the rat. Neurobiol Learn Mem. 2007;87:101–108. doi: 10.1016/j.nlm.2006.07.003. [DOI] [PubMed] [Google Scholar]
  241. Peters YM, O’Donnell P. Social isolation rearing affects prefrontal cortical response to ventral tegmental area stimulation. Biol Psychiatry. 2005;57:1205–1208. doi: 10.1016/j.biopsych.2005.02.011. [DOI] [PubMed] [Google Scholar]
  242. Pietropaolo S, Singer P, Feldon J, Yee BK. The postweaning social isolation in C57BL/6 mice: preferential vulnerability in the male sex. Psychopharmacology (Berl) 2008;197:613–628. doi: 10.1007/s00213-008-1081-3. [DOI] [PubMed] [Google Scholar]
  243. Piontkewitz Y, Assaf Y, Weiner I. Clozapine administration in adolescence prevents postpubertal emergence of brain structural pathology in an animal model of schizophrenia. Biol Psychiatry. 2009;66:1038–1046. doi: 10.1016/j.biopsych.2009.07.005. [DOI] [PubMed] [Google Scholar]
  244. Powell SB, Geyer MA. Developmental markers of psychiatric disorders as identified by sensorimotor gating. Neurotox Res. 2002;4:489–502. doi: 10.1080/10298420290030578. [DOI] [PubMed] [Google Scholar]
  245. Powell SB, Geyer MA. Overview of animal models of schizophrenia. Curr Protoc Neurosci. 2007;Chapter 9(Unit 9):24. doi: 10.1002/0471142301.ns0924s39. [DOI] [PubMed] [Google Scholar]
  246. Powell SB, Risbrough VB, Geyer MA. Potential use of animal models to examine antipsy-chotic prophylaxis for schizophrenia. Clin Neurosci Res. 2003;3:289–296. [Google Scholar]
  247. Powell SB, Zhou X, Geyer MA. Prepulse inhibition and genetic mouse models of schizophrenia. Behav Brain Res. 2009;204:282–294. doi: 10.1016/j.bbr.2009.04.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  248. Preece MA, Dalley JW, Theobald DE, Robbins TW, Reynolds GP. Region specific changes in forebrain 5-hydroxytryptamine1A and 5-hydroxytryptamine2A receptors in isolation-reared rats: an in vitro autoradiography study. Neuroscience. 2004;123:725–732. doi: 10.1016/j.neuroscience.2003.10.008. [DOI] [PubMed] [Google Scholar]
  249. Pryce CR, Bettschen D, Bahr NI, Feldon J. Comparison of the effects of infant handling, isolation, and nonhandling on acoustic startle, prepulse inhibition, locomotion, and HPA activity in the adult rat. Behav Neurosci. 2001;115:71–83. doi: 10.1037/0735-7044.115.1.71. [DOI] [PubMed] [Google Scholar]
  250. Ranade SC, Rose A, Rao M, Gallego J, Gressens P, Mani S. Different types of nutritional deficiencies affect different domains of spatial memory function checked in a radial arm maze. Neuroscience. 2008;152:859–866. doi: 10.1016/j.neuroscience.2008.01.002. [DOI] [PubMed] [Google Scholar]
  251. Rapoport JL, Addington AM, Frangou S, Psych MRC. The neurodevelopmental model of schizophrenia: update 2005. Mol Psychiatry. 2005;10:434–449. doi: 10.1038/sj.mp.4001642. [DOI] [PubMed] [Google Scholar]
  252. Rehn AE, Van Den Buuse M, Copolov D, Briscoe T, Lambert G, Rees S. An animal model of chronic placental insufficiency: relevance to neurodevelopmental disorders including schizophrenia. Neuroscience. 2004;129:381–391. doi: 10.1016/j.neuroscience.2004.07.047. [DOI] [PubMed] [Google Scholar]
  253. Reynolds GP, Beasley CL. GABAergic neuronal subtypes in the human frontal cortex –development and deficits in schizophrenia. J Chem Neuroanat. 2001;22:95–100. doi: 10.1016/s0891-0618(01)00113-2. [DOI] [PubMed] [Google Scholar]
  254. Reynolds GP, Abdul-Monim Z, Neill JC, Zhang ZJ. Calcium binding protein markers of GABA deficits in schizophrenia – postmortem studies and animal models. Neurotox Res. 2004;6:57–61. doi: 10.1007/BF03033297. [DOI] [PubMed] [Google Scholar]
  255. Rice JE, 3rd, Vannucci RC, Brierley JB. The influence of immaturity on hypoxic-ischemic brain damage in the rat. Ann Neurol. 1981;9:131–141. doi: 10.1002/ana.410090206. [DOI] [PubMed] [Google Scholar]
  256. Richtand NM, Taylor B, Welge JA, Ahlbrand R, Ostrander MM, Burr J, Hayes S, Coolen LM, Pritchard LM, Logue A, Herman JP, McNamara RK. Risperidone pretreatment prevents elevated locomotor activity following neonatal hippocampal lesions. Neuropsychopharmacology. 2006;31:77–89. doi: 10.1038/sj.npp.1300791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  257. Robbins TW. The 5-choice serial reaction time task: behavioural pharmacology and functional neurochemistry. Psychopharmacology (Berl) 2002;163:362–380. doi: 10.1007/s00213-002-1154-7. [DOI] [PubMed] [Google Scholar]
  258. Romero E, Ali C, Molina-Holgado E, Castellano B, Guaza C, Borrell J. Neurobehavioral and immunological consequences of prenatal immune activation in rats. Influence of antipsychotics. Neuropsychopharmacology. 2007;32:1791–1804. doi: 10.1038/sj.npp.1301292. [DOI] [PubMed] [Google Scholar]
  259. Romero E, Guaza C, Castellano B, Borrell J. Ontogeny of sensorimotor gating and immune impairment induced by prenatal immune challenge in rats: implications for the etiopathology of schizophrenia. Mol Psychiatry. 2008 doi: 10.1038/mp.2008.44. (Epub ahead of print, Apr 15) [DOI] [PubMed] [Google Scholar]
  260. Rothschild DM, O’Grady M, Wecker L. Neonatal cytomegalovirus exposure decreases prepulse inhibition in adult rats: implications for schizophrenia. J Neurosci Res. 1999;57:429–434. [PubMed] [Google Scholar]
  261. Rueter LE, Ballard ME, Gallagher KB, Basso AM, Curzon P, Kohlhaas KL. Chronic low dose risperidone and clozapine alleviate positive but not negative symptoms in the rat neonatal ventral hippocampal lesion model of schizophrenia. Psychopharmacology (Berl) 2004;176:312–319. doi: 10.1007/s00213-004-1897-4. [DOI] [PubMed] [Google Scholar]
  262. Sahakian BJ, Robbins TW, Morgan MJ, Iversen SD. The effects of psychomotor stimulants on stereotypy and locomotor activity in socially-deprived and control rats. Brain Res. 1975;84:195–205. doi: 10.1016/0006-8993(75)90975-0. [DOI] [PubMed] [Google Scholar]
  263. Sahakian BJ, Robbins TW, Iversen SD. The effects of isolation rearing on exploration in the rat. Anim Learn Behav. 1977;5:193–198. [Google Scholar]
  264. Sakaue M, Ago Y, Baba A, Matsuda T. The 5-HT1A receptor agonist MKC-242 reverses isolation rearing-induced deficits of prepulse inhibition in mice. Psychopharmacology (Berl) 2003;170:73–79. doi: 10.1007/s00213-003-1515-x. [DOI] [PubMed] [Google Scholar]
  265. Sams-Dodd F. Phencyclidine-induced stereotyped behaviour and social isolation in rats: a possible animal model of schizophrenia. Behav Pharmacol. 1996;7:3–23. [PubMed] [Google Scholar]
  266. Sams-Dodd F. A test of the predictive validity of animal models of schizophrenia based on phencyclidine and D-amphetamine. Neuropsychopharmacology. 1998;18:293–304. doi: 10.1016/S0893-133X(97)00161-9. [DOI] [PubMed] [Google Scholar]
  267. Sams-Dodd F, Lipska BK, Weinberger DR. Neonatal lesions of the rat ventral hippocampus result in hyperlocomotion and deficits in social behaviour in adulthood. Psychopharmacology (Berl) 1997;132:303–310. doi: 10.1007/s002130050349. [DOI] [PubMed] [Google Scholar]
  268. Sandager-Nielsen K, Andersen MB, Sager TN, Werge T, Scheel-Kruger J. Effects of postnatal anoxia on striatal dopamine metabolism and prepulse inhibition in rats. Pharmacol Biochem Behav. 2004;77:767–774. doi: 10.1016/j.pbb.2004.01.017. [DOI] [PubMed] [Google Scholar]
  269. Sarter M, Givens B, Bruno JP. The cognitive neuroscience of sustained attention: where top-down meets bottom-up. Brain Res Brain Res Rev. 2001;35:146–160. doi: 10.1016/s0165-0173(01)00044-3. [DOI] [PubMed] [Google Scholar]
  270. Scaccianoce S, Del Bianco P, Paolone G, Caprioli D, Modafferi AM, Nencini P, Badiani A. Social isolation selectively reduces hippocampal brain-derived neurotrophic factor without altering plasma corticosterone. Behav Brain Res. 2006;168:323–325. doi: 10.1016/j.bbr.2005.04.024. [DOI] [PubMed] [Google Scholar]
  271. Schiavone S, Sorce S, Dubois-Dauphin M, Jaquet V, Colaianna M, Zotti M, Cuomo V, Trabace L, Krause KH. Involvement of NOX2 in the development of behavioral and pathologic alterations in isolated rats. Biol Psychiatry. 2009;66:384–392. doi: 10.1016/j.biopsych.2009.04.033. [DOI] [PubMed] [Google Scholar]
  272. Schrijver NC, Wurbel H. Early social deprivation disrupts attentional, but not affective, shifts in rats. Behav Neurosci. 2001;115:437–442. [PubMed] [Google Scholar]
  273. Schrijver NC, Pallier PN, Brown VJ, Wurbel H. Double dissociation of social and environmental stimulation on spatial learning and reversal learning in rats. Behav Brain Res. 2004;152:307–314. doi: 10.1016/j.bbr.2003.10.016. [DOI] [PubMed] [Google Scholar]
  274. Schubert MI, Porkess MV, Dashdorj N, Fone KC, Auer DP. Effects of social isolation rearing on the limbic brain: a combined behavioral and magnetic resonance imaging volumetry study in rats. Neuroscience. 2009;159:21–30. doi: 10.1016/j.neuroscience.2008.12.019. [DOI] [PubMed] [Google Scholar]
  275. Schwarz MJ, Muller N, Riedel M, Ackenheil M. The Th2-hypothesis of schizophrenia: a strategy to identify a subgroup of schizophrenia caused by immune mechanisms. Med Hypotheses. 2001;56:483–486. doi: 10.1054/mehy.2000.1203. [DOI] [PubMed] [Google Scholar]
  276. Secher T, Berezin V, Bock E, Glenthoj B. Effect of an NCAM mimetic peptide FGL on impairment in spatial learning and memory after neonatal phencyclidine treatment in rats. Behav Brain Res. 2009;199:288–297. doi: 10.1016/j.bbr.2008.12.012. [DOI] [PubMed] [Google Scholar]
  277. Segal DS, Geyer MA. Animal models of psychopathology. In: Judd LL, Groves PM, editors. Psychobiological foundations of clinical psychiatry. JB Lippincott Co; Philadelphia: 1985. pp. 1–14. [Google Scholar]
  278. Segal DS, Geyer MA, Schuckit A. Stimulant-induced psychosis: an evaluation of animal models. In: Youdim MBH, Lovenberg W, Sharman DF, Lagnado JR, editors. Essays in neurochemistry and neuropharmacology. Wiley; New York: 1981. pp. 95–130. [PubMed] [Google Scholar]
  279. Selten JP, Brown AS, Moons KG, Slaets JP, Susser ES, Kahn RS. Prenatal exposure to the 1957 influenza pandemic and non-affective psychosis in The Netherlands. Schizophr Res. 1999a;38:85–91. doi: 10.1016/s0920-9964(99)00005-5. [DOI] [PubMed] [Google Scholar]
  280. Selten JP, van der Graaf Y, van Duursen R, Gispen-de Wied CC, Kahn RS. Psychotic illness after prenatal exposure to the 1953 Dutch flood disaster. Schizophr Res. 1999b;35:243–245. doi: 10.1016/s0920-9964(98)00143-1. [DOI] [PubMed] [Google Scholar]
  281. Shi L, Fatemi SH, Sidwell RW, Patterson PH. Maternal influenza infection causes marked behavioral and pharmacological changes in the offspring. J Neurosci. 2003;23:297–302. doi: 10.1523/JNEUROSCI.23-01-00297.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  282. Silva-Gomez AB, Bermudez M, Quirion R, Srivastava LK, Picazo O, Flores G. Comparative behavioral changes between male and female postpubertal rats following neonatal excitotoxic lesions of the ventral hippocampus. Brain Res. 2003a;973:285–292. doi: 10.1016/s0006-8993(03)02537-x. [DOI] [PubMed] [Google Scholar]
  283. Silva-Gomez AB, Rojas D, Juarez I, Flores G. Decreased dendritic spine density on prefrontal cortical and hippocampal pyramidal neurons in postweaning social isolation rats. Brain Res. 2003b;983:128–136. doi: 10.1016/s0006-8993(03)03042-7. [DOI] [PubMed] [Google Scholar]
  284. Sircar R, Rudy JW. Repeated neonatal phencyclidine treatment impairs performance of a spatial task in juvenile rats. Ann N Y Acad Sci. 1998;844:303–309. [PubMed] [Google Scholar]
  285. Smith SEP, Li J, Garbett K, Mirnics K, Patterson PH. Maternal immune activation alters fetal brain development through interleukin-6. J Neurosci. 2007;27:10695–10702. doi: 10.1523/JNEUROSCI.2178-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  286. Son GH, Geum D, Chung S, Kim EJ, Jo JH, Kim CM, Lee KH, Kim H, Choi S, Kim HT, Lee CJ, Kim K. Maternal stress produces learning deficits associated with impairment of NMDA receptor-mediated synaptic plasticity. J Neurosci. 2006;26:3309–3318. doi: 10.1523/JNEUROSCI.3850-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  287. Son GH, Chung S, Geum D, Kang SS, Choi WS, Kim K, Choi S. Hyperactivity and alteration of the midbrain dopaminergic system in maternally stressed male mice offspring. Biochem Biophys Res Commun. 2007;352:823–829. doi: 10.1016/j.bbrc.2006.11.104. [DOI] [PubMed] [Google Scholar]
  288. 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. doi: 10.1093/schbul/sbn121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  289. Stefani MR, Moghaddam B. Transient N-methyl-D-aspartate receptor blockade in early development causes lasting cognitive deficits relevant to schizophrenia. Biol Psychiatry. 2005;57:433–436. doi: 10.1016/j.biopsych.2004.11.031. [DOI] [PubMed] [Google Scholar]
  290. Stevens KE, Johnson RG, Rose GM. Rats reared in social isolation show schizophrenia-like changes in auditory gating. Pharmacol Biochem Behav. 1997;58:1031–1036. doi: 10.1016/s0091-3057(97)00306-7. [DOI] [PubMed] [Google Scholar]
  291. Susser E, Neugebauer R, Hoek HW, Brown AS, Lin S, Labovitz D, Gorman JM. Schizophrenia after prenatal famine: further evidence. Arch Gen Psychiatry. 1996;53:25–31. doi: 10.1001/archpsyc.1996.01830010027005. [DOI] [PubMed] [Google Scholar]
  292. Swerdlow NR, Geyer MA. Using an animal model of deficient sensorimotor gating to study the pathophysiology and new treatments of schizophrenia. Schizophr Bull. 1998;24:285–301. doi: 10.1093/oxfordjournals.schbul.a033326. [DOI] [PubMed] [Google Scholar]
  293. Swerdlow NR, Vaccarino FJ, Amalric M, Koob GF. The neural substrates for the motor-activating properties of psychostimulants: a review of recent findings. Pharmacol Biochem Behav. 1986;25:233–248. doi: 10.1016/0091-3057(86)90261-3. [DOI] [PubMed] [Google Scholar]
  294. Swerdlow NR, Braff DL, Taaid N, Geyer MA. Assessing the validity of an animal model of deficient sensorimotor gating in schizophrenic patients. Arch Gen Psychiatry. 1994;51:139–154. doi: 10.1001/archpsyc.1994.03950020063007. [DOI] [PubMed] [Google Scholar]
  295. Swerdlow NR, Light GA, Breier M, Ko D, Mora AB, Shoemaker JM, Saint Marie RL, Neary AC, Geyer MA, Stevens KE, Powell SB. Sensory and sensorimotor gating deficits after neonatal ventral hippocampal lesions in rats. 37th Annual meeting of the society for neuroscience; San Diego, CA. 2007. p. Abstract No. 607.9. [DOI] [PubMed] [Google Scholar]
  296. Swerdlow NR, Weber M, Qu Y, Light GA, Braff DL. Realistic expectations of prepulse inhibition in translational models for schizophrenia research. Psychopharmacology (Berl) 2008;199:331–388. doi: 10.1007/s00213-008-1072-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  297. Szuran TF, Pliska V, Pokorny J, Welzl H. Prenatal stress in rats: effects on plasma corticosterone, hippocampal glucocorticoid receptors, and maze performance. Physiol Behav. 2000;71:353–362. doi: 10.1016/s0031-9384(00)00351-6. [DOI] [PubMed] [Google Scholar]
  298. Takei N, Lewis S, Jones P, Harvey I, Murray RM. Prenatal exposure to influenza and increased cerebrospinal fluid spaces in schizophrenia. Schizophr Bull. 1996;22:521–534. doi: 10.1093/schbul/22.3.521. [DOI] [PubMed] [Google Scholar]
  299. Talamini LM, Koch T, Ter Horst GJ, Korf J. Methylazoxymethanol acetate-induced abnormalities in the entorhinal cortex of the rat; parallels with morphological findings in schizophrenia. Brain Res. 1998;789:293–306. doi: 10.1016/s0006-8993(98)00020-1. [DOI] [PubMed] [Google Scholar]
  300. Talamini LM, Koch T, Luiten PG, Koolhaas JM, Korf J. Interruptions of early cortical development affect limbic association areas and social behaviour in rats; possible relevance for neurodevelopmental disorders. Brain Res. 1999;847:105–120. doi: 10.1016/s0006-8993(99)02067-3. [DOI] [PubMed] [Google Scholar]
  301. Talamini LM, Ellenbroek B, Koch T, Korf J. Impaired sensory gating and attention in rats with developmental abnormalities of the mesocortex. Implications for schizophrenia. Ann N Y Acad Sci. 2000;911:486–494. doi: 10.1111/j.1749-6632.2000.tb06751.x. [DOI] [PubMed] [Google Scholar]
  302. Thor DH, Holloway WR. Persistence of social investigatory behavior in the male rat: evidence for long-term memory of initial copluatory experience. Anim Learn Behav. 1981;9:561–565. [Google Scholar]
  303. Tohmi M, Tsuda N, Watanabe Y, Kakita A, Nawa H. Perinatal inflammatory cytokine challenge results in distinct neurobehavioral alterations in rats: implication in psychiatric disorders of developmental origin. Neurosci Res. 2004;50:67–75. doi: 10.1016/j.neures.2004.05.010. [DOI] [PubMed] [Google Scholar]
  304. Tonkiss J, Galler JR. Prenatal protein malnutrition and working memory performance in adult rats. Behav Brain Res. 1990;40:95–107. doi: 10.1016/0166-4328(90)90002-v. [DOI] [PubMed] [Google Scholar]
  305. Tonkiss J, Almeida SS, Galler JR. Prenatally malnourished female but not male rats show increased sensitivity to MK-801 in a differential reinforcement of low rates task. Behav Pharmacol. 1998;9:49–60. [PubMed] [Google Scholar]
  306. Torrey EF, Miller J, Rawlings R, Yolken RH. Seasonality of births in schizophrenia and bipolar disorder: a review of the literature. Schizophr Res. 1997;28:1–38. doi: 10.1016/s0920-9964(97)00092-3. [DOI] [PubMed] [Google Scholar]
  307. Tseng KY, O’Donnell P. Dopamine modulation of prefrontal cortical interneurons changes during adolescence. Cereb Cortex. 2007;17:1235–1240. doi: 10.1093/cercor/bhl034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  308. Tseng KY, Lewis BL, Lipska BK, O’Donnell P. Post-pubertal disruption of medial prefrontal cortical dopamine–glutamate interactions in a developmental animal model of schizophrenia. Biol Psychiatry. 2007;62:730–738. doi: 10.1016/j.biopsych.2006.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  309. Tseng KY, Lewis BL, Hashimoto T, Sesack SR, Kloc M, Lewis DA, O’Donnell P. A neonatal ventral hippocampal lesion causes functional deficits in adult prefrontal cortical interneurons. J Neurosci. 2008;28:12691–12699. doi: 10.1523/JNEUROSCI.4166-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  310. Tseng KY, Chambers RA, Lipska BK. The neonatal ventral hippocampal lesion as a heuristic neurodevelopmental model of schizophrenia. Behav Brain Res. 2009;204:295–305. doi: 10.1016/j.bbr.2008.11.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  311. Tsuda N, Tohmi M, Mizuno M, Nawa H. Strain-dependent behavioral alterations induced by peripheral interleukin-1 challenge in neonatal mice. Behav Brain Res. 2006;166:19–31. doi: 10.1016/j.bbr.2005.07.007. [DOI] [PubMed] [Google Scholar]
  312. Urakubo A, Jarskog LF, Lieberman JA, Gilmore JH. Prenatal exposure to maternal infection alters cytokine expression in the placenta, amniotic fluid, and fetal brain. Schizophr Res. 2001;47:27–36. doi: 10.1016/s0920-9964(00)00032-3. [DOI] [PubMed] [Google Scholar]
  313. Vaillancourt C, Boksa P. Birth insult alters dopamine-mediated behavior in a precocial species, the guinea pig. Implications for schizophrenia. Neuropsychopharmacology. 2000;23:654–666. doi: 10.1016/S0893-133X(00)00164-0. [DOI] [PubMed] [Google Scholar]
  314. van Os J, Selten JP. Prenatal exposure to maternal stress and subsequent schizophrenia. The May 1940 invasion of The Netherlands. Br J Psychiatry. 1998;172:324–326. doi: 10.1192/bjp.172.4.324. [DOI] [PubMed] [Google Scholar]
  315. Varty GB, Geyer MA. Effects of isolation rearing on startle reactivity, habituation, and prepulse inhibition in male Lewis, Sprague-Dawley, and Fischer F344 rats. Behav Neurosci. 1998;112:1450–1457. doi: 10.1037//0735-7044.112.6.1450. [DOI] [PubMed] [Google Scholar]
  316. Varty GB, Higgins GA. Examination of drug-induced and isolation-induced disruptions of prepulse inhibition as models to screen antipsychotic drugs. Psychopharmacology (Berl) 1995;122:15–26. doi: 10.1007/BF02246437. [DOI] [PubMed] [Google Scholar]
  317. Varty GB, Marsden CA, Higgins GA. Reduced synaptophysin immunoreactivity in the dentate gyrus of prepulse inhibition-impaired isolation-reared rats. Brain Res. 1999;824:197–203. doi: 10.1016/s0006-8993(99)01173-7. [DOI] [PubMed] [Google Scholar]
  318. Varty GB, Paulus MP, Braff DL, Geyer MA. Environmental enrichment and isolation rearing in the rat: effects on locomotor behavior and startle response plasticity. Biol Psychiatry. 2000;47:864–873. doi: 10.1016/s0006-3223(99)00269-3. [DOI] [PubMed] [Google Scholar]
  319. Varty GB, Powell SB, Lehmann-Masten V, Buell MR, Geyer MA. Isolation rearing of mice induces deficits in prepulse inhibition of the startle response. Behav Brain Res. 2006;169:162–167. doi: 10.1016/j.bbr.2005.11.025. [DOI] [PubMed] [Google Scholar]
  320. Vohs JL, Chambers RA, Krishnan GP, O’Donnell BF, Hetrick WP, Kaiser ST, Berg S, Morzorati SL. Auditory sensory gating in the neonatal ventral hippocampal lesion model of schizophrenia. Neuropsychobiology. 2009;60:12–22. doi: 10.1159/000234813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  321. Voikar V, Polus A, Vasar E, Rauvala H. Long-term individual housing in C57BL/6J and DBA/2 mice: assessment of behavioral consequences. Genes Brain Behav. 2005;4:240–252. doi: 10.1111/j.1601-183X.2004.00106.x. [DOI] [PubMed] [Google Scholar]
  322. Wakuda T, Matsuzaki H, Suzuki K, Iwata Y, Shinmura C, Suda S, Iwata K, Yamamoto S, Sugihara G, Tsuchiya KJ, Ueki T, Nakamura K, Nakahara D, Takei N, Mori N. Perinatal asphyxia reduces dentate granule cells and exacerbates methamphetamine-induced hyperlocomotion in adulthood. PLoS One. 2008;3:e3648. doi: 10.1371/journal.pone.0003648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  323. Wang C, McInnis J, West JB, Bao J, Anastasio N, Guidry JA, Ye Y, Salvemini D, Johnson KM. Blockade of phencyclidine-induced cortical apoptosis and deficits in prepulse inhibition by M40403, a superoxide dismutase mimetic. J Pharmacol Exp Ther. 2003;304:266–271. doi: 10.1124/jpet.102.041798. [DOI] [PubMed] [Google Scholar]
  324. Wang CZ, Yang SF, Xia Y, Johnson KM. Postnatal phencyclidine administration selectively reduces adult cortical parvalbumin-containing interneurons. Neuropsychopharmacology. 2008;33:2442–2455. doi: 10.1038/sj.npp.1301647. [DOI] [PubMed] [Google Scholar]
  325. Watanabe Y, Hashimoto S, Kakita A, Takahashi H, Ko J, Mizuno M, Someya T, Patterson PH, Nawa H. Neonatal impact of leukemia inhibitory factor on neurobehavioral development in rats. Neurosci Res. 2004;48:345–353. doi: 10.1016/j.neures.2003.12.001. [DOI] [PubMed] [Google Scholar]
  326. Wedzony K, Fijal K, Mackowiak M, Chocyk A, Zajaczkowski W. Impact of postnatal blockade of N-methyl-D-aspartate receptors on rat behavior: a search for a new developmental model of schizophrenia. Neuroscience. 2008;153:1370–1379. doi: 10.1016/j.neuroscience.2008.03.016. [DOI] [PubMed] [Google Scholar]
  327. Weinberger DR. Implications of normal brain development for the pathogenesis of schizophrenia. Arch Gen Psychiatry. 1987;44:660–669. doi: 10.1001/archpsyc.1987.01800190080012. [DOI] [PubMed] [Google Scholar]
  328. Weinberger DR, Berman KF, Suddath R, Torrey EF. Evidence of dysfunction of a prefrontal-limbic network in schizophrenia: a magnetic resonance imaging and regional cerebral blood flow study of discordant monozygotic twins. Am J Psychiatry. 1992;149:890–897. doi: 10.1176/ajp.149.7.890. [DOI] [PubMed] [Google Scholar]
  329. Weiner I, Arad M. Using the pharmacology of latent inhibition to model domains of pathology in schizophrenia and their treatment. Behav Brain Res. 2009;204:369–386. doi: 10.1016/j.bbr.2009.05.004. [DOI] [PubMed] [Google Scholar]
  330. Weiner I, Schnabel I, Lubow RE, Feldon J. The effects of early handling on latent inhibition in male and female rats. Dev Psychobiol. 1985;18:291–297. doi: 10.1002/dev.420180402. [DOI] [PubMed] [Google Scholar]
  331. Weiner I, Lubow RE, Feldon J. Disruption of latent inhibition by acute administration of low doses of amphetamine. Pharmacol Biochem Behav. 1988;30:871–878. doi: 10.1016/0091-3057(88)90113-x. [DOI] [PubMed] [Google Scholar]
  332. Weinstock M. The long-term behavioural consequences of prenatal stress. Neurosci Bio-behav Rev. 2008;32:1073–1086. doi: 10.1016/j.neubiorev.2008.03.002. [DOI] [PubMed] [Google Scholar]
  333. Weiss IC, Feldon J. Environmental animal models for sensorimotor gating deficiencies in schizophrenia: a review. Psychopharmacology (Berl) 2001;156:305–326. doi: 10.1007/s002130100800. [DOI] [PubMed] [Google Scholar]
  334. Weiss IC, Domeney AM, Moreau JL, Russig H, Feldon J. Dissociation between the effects of pre-weaning and/or post-weaning social isolation on prepulse inhibition and latent inhibition in adult Sprague-Dawley rats. Behav Brain Res. 2001;121:207–218. doi: 10.1016/s0166-4328(01)00166-8. [DOI] [PubMed] [Google Scholar]
  335. Weiss IC, Pryce CR, Jongen-Relo AL, Nanz-Bahr NI, Feldon J. Effect of social isolation on stress-related behavioural and neuroendocrine state in the rat. Behav Brain Res. 2004;152:279–295. doi: 10.1016/j.bbr.2003.10.015. [DOI] [PubMed] [Google Scholar]
  336. Wiley JL, Buhler KG, Lavecchia KL, Johnson KM. Pharmacological challenge reveals long-term effects of perinatal phencyclidine on delayed spatial alternation in rats. Prog Neuropsychopharmacol Biol Psychiatry. 2003;27:867–873. doi: 10.1016/S0278-5846(03)00146-5. [DOI] [PubMed] [Google Scholar]
  337. Wilkinson LS, Killcross SS, Humby T, Hall FS, Geyer MA, Robbins TW. Social isolation in the rat produces developmentally specific deficits in prepulse inhibition of the acoustic startle response without disrupting latent inhibition. Neuropsychopharmacology. 1994;10:61–72. doi: 10.1038/npp.1994.8. [DOI] [PubMed] [Google Scholar]
  338. Winocur G. Functional dissociation of the hippocampus and prefrontal cortex in learning and memory. Psychobiology. 1991;19:11–20. [Google Scholar]
  339. Winslow JT, Camacho F. Cholinergic modulation of a decrement in social investigation following repeated contacts between mice. Psychopharmacology (Berl) 1995;121:164–172. doi: 10.1007/BF02245626. [DOI] [PubMed] [Google Scholar]
  340. Winter C, Djodari-Irani A, Sohr R, Morgenstern R, Feldon J, Juckel G, Meyer U. Prenatal immune activation leads to multiple changes in basal neurotransmitter levels in the adult brain: implications for brain disorders of neurodevelopmental origin such as schizophrenia. Int J Neuropsychopharmacol. 2009;12:513–524. doi: 10.1017/S1461145708009206. [DOI] [PubMed] [Google Scholar]
  341. Wolff AR, Bilkey DK. Immune activation during mid-gestation disrupts sensorimotor gating in rat offspring. Behav Brain Res. 2008;190:156–159. doi: 10.1016/j.bbr.2008.02.021. [DOI] [PubMed] [Google Scholar]
  342. Wong AH, Lipska BK, Likhodi O, Boffa E, Weinberger DR, Kennedy JL, Van Tol HH. Cortical gene expression in the neonatal ventral–hippocampal lesion rat model. Schizophr Res. 2005;77:261–270. doi: 10.1016/j.schres.2005.03.011. [DOI] [PubMed] [Google Scholar]
  343. Wright IK, Upton N, Marsden CA. Resocialisation of isolation-reared rats does not alter their anxiogenic profile on the elevated X-maze model of anxiety. Physiol Behav. 1991;50:1129–1132. doi: 10.1016/0031-9384(91)90572-6. [DOI] [PubMed] [Google Scholar]
  344. Wu J, Song TB, Li YJ, He KS, Ge L, Wang LR. Prenatal restraint stress impairs learning and memory and hippocampal PKCbeta1 expression and translocation in offspring rats. Brain Res. 2007;1141:205–213. doi: 10.1016/j.brainres.2007.01.024. [DOI] [PubMed] [Google Scholar]
  345. Xu M-Q, Sun W-S, Liu B-X, Feng G-Y, Yu L, Yang L, He G, Sham P, Susser E, St Clair D, He L. Prenatal malnutrition and adult schizophrenia: further evidence from the 1959–1961 Chinese famine. Schizophr Bull. 2009;35:568–576. doi: 10.1093/schbul/sbn168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  346. Yang J, Han H, Cao J, Li L, Xu L. Prenatal stress modifies hippocampal synaptic plasticity and spatial learning in young rat offspring. Hippocampus. 2006;16:431–436. doi: 10.1002/hipo.20181. [DOI] [PubMed] [Google Scholar]
  347. Young JW, Powell SB, Risbrough V, Marston HM, Geyer MA. Using the MATRICS to guide development of a preclinical cognitive test battery for research in schizophrenia. Pharmacol Ther. 2009;122:150–202. doi: 10.1016/j.pharmthera.2009.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  348. Young JW, Zhou X, Geyer MA. Animal models of schizophrenia. Behavioral Neurobiology of Schizophrenia and Its Treatment. In: Swerdlow N, editor. Curr Topics in Behav Neurosci. Springer; Heidelberg: 2010. [DOI] [PubMed] [Google Scholar]
  349. Zornberg GL, Buka SL, Tsuang MT. The problem of obstetrical complications and schizophrenia. Schizophr Bull. 2000;26:249–256. doi: 10.1093/oxfordjournals.schbul.a033449. [DOI] [PubMed] [Google Scholar]
  350. Zorrilla EP. Multiparous species present problems (and possibilities) to developmentalists. Dev Psychobiol. 1997;30:141–150. doi: 10.1002/(sici)1098-2302(199703)30:2<141::aid-dev5>3.0.co;2-q. [DOI] [PubMed] [Google Scholar]
  351. Zuckerman L, Weiner I. Post-pubertal emergence of disrupted latent inhibition following prenatal immune activation. Psychopharmacology (Berl) 2003;169:308–313. doi: 10.1007/s00213-003-1461-7. [DOI] [PubMed] [Google Scholar]
  352. Zuckerman L, Weiner I. Maternal immune activation leads to behavioral and pharmacological changes in the adult offspring. J Psychiatr Res. 2005;39:311–323. doi: 10.1016/j.jpsychires.2004.08.008. [DOI] [PubMed] [Google Scholar]
  353. Zuckerman L, Rehavi M, Nachman R, Weiner I. Immune activation during pregnancy in rats leads to a postpubertal emergence of disrupted latent inhibition, dopaminergic hyperfunction, and altered limbic morphology in the offspring: a novel neurodevelopmental model of schizophrenia. Neuropsychopharmacology. 2003;28:1778–1789. doi: 10.1038/sj.npp.1300248. [DOI] [PubMed] [Google Scholar]

RESOURCES