Abstract
Genetic and environmental models of neuropsychiatric disease have grown exponentially over the last 20 years. One measure that is often used to evaluate the translational relevance of these models to human neuropsychiatric disease is prepulse inhibition of startle (PPI), an operational measure of sensorimotor gating. Deficient PPI characterizes several neuropsychiatric disorders but has been most extensively studied in schizophrenia. It has become a useful tool in translational neuropharmacological and molecular genetics studies because it can be measured across species using almost the same experimental parameters. Although initial studies of PPI in rodents were pharmacological because of the robust predictive validity of PPI for antipsychotic efficacy, more recently, PPI has become standard common behavioral measures used in genetic and neurodevelopmental models of schizophrenia. Here we review “two hit” models of schizophrenia and discuss the utility of PPI as a tool in phenotyping these models of relevant risk factors. In the review, we consider approaches to rodent models of genetic and neurodevelopmental risk factors and selectively review “two hit” models of gene × environment and environment × environment interactions in which PPI has been measured.
Keywords: behavior, development, prepulse inhibition, schizophrenia, genetic, risk factor, double hit, sensorimotor gating
1. Introduction: Utility of Prepulse Inhibition in Models Relevant to Schizophrenia
Sensorimotor gating occurs when a motor responses is gated by a sensory event. One form of sensorimotor gating that has been studied at multiple levels of biology, from its cellular mechanisms (Frost et al., 2003; Nusbaum and Contreras, 2004; Rose and Scott, 2003) to its relationship to neuropsychiatric disease (Braff, 2010, 2011; Swerdlow et al., 2008), is prepulse inhibition (PPI) of startle. PPI occurs when a weak, subthreshold stimulus presented 30–500 ms prior to an intense startling stimulus inhibits the startle response (Graham, 1975; Hoffman and Ison, 1980). The circuitry of PPI has been studied most extensively in rodents and involves role of cortico-striatal-pedunculo-pontine (CSPP) circuitry in which limbic and descending pontine projections modulate the ability of the prepulse to inhibit the startle response, which occurs at the level of the pons (Swerdlow et al., 2001a; Swerdlow et al., 2008). Thus, PPI provides an operational measure of sensorimotor gating and may indicate the integrity of the underlying neural circuitry subserving sensorimotor gating mechanisms. PPI is an integral part of human psychophysiological studies of neuropsychiatric disease and is amenable to neuroscience-based inquiry of deficits in functional domains. Indeed, in the Research Domain Criteria (RDoC) outlined by the National Institute of Mental Health, PPI is considered part of the “Auditory Perception” construct in the cognitive domain. In humans, startle to acoustic or tactile stimuli is most often measured from the eye blink response (Braff et al., 1992; Fridlund and Cacioppo, 1986; Kumari et al., 2003; Neuner et al., 2010; Swerdlow et al., 2001b). PPI deficits were first observed in schizophrenia patients (for review see Braff et al., 2001; Swerdlow et al., 2014; Swerdlow et al., 2008), but are also apparent in their unaffected first degree relatives (Cadenhead et al., 2000) as well as patients with schizotypal personality disorder (Cadenhead et al., 1993). A recent large, multi-site study reported PPI deficits in schizophrenia patients, corroborating the more than 40 single-site studies published to date (Swerdlow et al., 2014). PPI deficits, however, are not unique to schizophrenia and are also observed in several other neuropsychiatric disorders (Kohl et al., 2013), including Obsessive-Compulsive Disorder (Ahmari et al., 2012; Ahmari et al., 2016; Hoenig et al., 2005; Swerdlow et al., 1993), Tourette’s syndrome (Buse et al., 2016; Castellanos et al., 1996; Swerdlow et al., 2001b), Huntington’s disease (Swerdlow et al., 1995; Valls-Sole et al., 2004), manic bipolar patients (Perry et al., 2001), Panic Disorder (Ludewig et al., 2002), Fragile × syndrome (Frankland et al., 2004; Hessl et al., 2009), adults with autism (Perry et al., 2007), Asperger’s Syndrome (McAlonan et al., 2002), 22q11 Syndrome (Sobin et al., 2005), nocturnal enuresis (Ornitz et al., 1992), and Kleinfelter Syndrome (van Rijn et al., 2011). Thus, PPI deficits are observed across many neuropsychiatric disorders but have been the most widely replicated in schizophrenia patients (Braff et al., 2001; Kumari et al., 2008; Ludewig et al., 2003; Mackeprang et al., 2002; Swerdlow et al., 2008).
PPI has been a useful behavioral phenotype to consider in genetic mouse models relevant to schizophrenia and other neuropsychiatric diseases (Powell et al., 2012). Additionally, because PPI measures basic information processing and can be quantified in multiple species, it is a useful tool for understanding the biology of putative risk genes. Indeed, over the last 20 years a large number of genetic mouse models have been tested for differences in PPI. These studies indicate that PPI can be either increased or decreased by a wide variety genes involved in neural development, neurotransmitter function, or basic cellular processes (Powell et al. 2009; Powell et al. 2012). Our recent review provided an update on mutant mouse models in which PPI was measured as a phenotype with comprehensive tables detailing PPI across a wide variety of mutant models and its pharmacological modulation, where appropriate (Powell et al., 2012). PPI has also proven to be a useful tool in evaluating the impact of environmental risk factors during development, which is covered briefly in Section 3.
Previous reviews summarized schizophrenia candidate genes (Arguello and Gogos, 2010; Arguello and Gogos, 2011; O’Tuathaigh and Waddington, 2015), while other reviews focused specifically on PPI, summarizing genetic mutants, strain differences, and the pharmacology of PPI in mice (Geyer et al., 2002; Powell et al., 2012; Powell et al., 2009; Swerdlow et al., 2008; van den Buuse, 2010), as well as recent reviews on models of gene × environment interactions (Ayhan et al., 2016; Moran et al., 2016). The etiology of schizophrenia is multifaceted and likely involves a convergence of both genetic and environmental risk factors (Cannon et al., 2003; Gottesman, 1991; Uher, 2014). Thus, experimental models evaluating gene-environment interactions are particularly informative for schizophrenia. In this review, we summarize approaches to rodent models of genetic and neurodevelopmental risk factors and selectively review “two hit” models of gene × environment and environment × environment interactions in which PPI has been measured. The review highlights approaches to combined risk factors for schizophrenia that have used PPI as a behavioral endpoint and discusses caveats of, and future directions for, double hit models.
2. Genetic Landscape of Schizophrenia
2.1 Approaches to genetic discoveries
The two primary approaches to understanding the genetics of neuropsychiatric disease are the common disease / common allele approach (CDCA) and the common disease / rare allele approach (CDRA) (Arguello and Gogos, 2011). Candidate gene or unbiased genome-wide association studies (GWAS) focus on common genetic variants (>5% allele frequency); whereas, the CDRA approach focuses on the hypothesis that rare variants with high penetrance can cause common disease (Arguello and Gogos, 2011). Schizophrenia and other major neuropsychiatric and neurodevelopmental conditions are likely a combination of risk from both common and rare variants.
The recent Psychiatric Genomics Consortium (PGC) genome-wide association study (GWAS) of schizophrenia (Consortium, 2014) identified 108 genetic loci associated with schizophrenia. Some of the most notable findings in the PGC are loci containing genes for G protein coupled receptor signaling, glutamate neurotransmission, neuronal calcium signaling, synaptic function and plasticity, other neuronal ion channels, and neurodevelopment (Consortium, 2014). Because these associations imply the existence of one or more risk variants at the locus rather than a specific gene, it is premature to discuss in depth the role of any specific genes at these loci until there is a more complete understanding of the risk variants and whether the variants are functional. As the basic biology of the identified loci begins to be investigated, there will certainly be many mouse mutants created to target those genes. One strategy for using the PGC GWAS data for neuroscience drug discovery put forth by Schubert and colleagues, is to prioritize gene targets based on knowledge of gene function and functional variants to identify putatively causal genes, and annotate these putatively causal genes with information on mRNA expression, de novo mutations, disease-associated rare mutations, and literature knowledge to determine targets for novel drug discovery (Schubert et al., 2014). A similar strategy could be taken by molecular biologists creating novel mouse mutants for basic biological interrogations of target genes. Another interesting finding emerging from large-scale GWAS studies across psychiatric disorders is the large degree of genetic overlap between schizophrenia and both autism spectrum disorder (ASD) and bipolar disorder, suggesting shared disease pathways or common risk. Thus, mouse models manipulating these genes should be considered a more general risk factor for multiple neurodevelopmental and/or neuropsychiatric disorders.
2.2 Genetics of PPI as an endophenotype
A complementary approach to large-scale GWAS or copy number variant (CNV) studies of schizophrenia are genetic studies of endophenotypes, which assume that the endophenotypemore proximal to the biological function of disrupted genes and/or be more easily and reliably quantified. Hence, psychophysiological processes such as PPI, have been used as endophenotypes in schizophrenia genetic studies (Braff et al., 2007; Greenwood et al., 2011; Greenwood et al., 2012; Greenwood et al., 2013) based on meeting criteria for a viable endophenotype (e.g. heritable, easily measured, good test-retest reliability; (Turetsky et al., 2007). PPI heritability has been estimated at 32%, which is similar to the 31% and 44% schizophrenia heritability estimates for nuclear and extended families, respectively, suggesting similar heritabilities for the disease and the endophenotype (Greenwood et al., 2007; Light et al., 2014).
Candidate gene studies indicated that polymorphisms in the CHRNA3 gene (Petrovsky et al., 2010), neuregulin 1 (Roussos et al., 2011), and COMT (Giakoumaki et al., 2008; Quednow et al., 2008; Roussos et al., 2008) are associated with PPI. In more recent studies of multiple SNPs using much larger sample sizes, however, only a few of these associations remained. In the larger, family-based COGS (Consortium on the Genetics of Schizophrenia) dataset, SNPs for CHRNA7, NCAM1, COMT, GRID2, CAMK2A were the most strongly associated with PPI, and NOS1AP, GRIK3, NRG1, GRIN3A, and DBH moderately associated with PPI (Greenwood et al., 2011). In a follow-up study based on non-familial samples from UCSD (Greenwood et al., 2012) only GRID2 achieved significance at more stringent significance levels. Other genes including GRIK3, CTNNA2, SLC6A3, SLC1A2, and GRIN2A were modestly associated with PPI. Across the endophenotypes studied in the UCSD and COGS samples, GRID2 and GRIK3 were significantly associated with PPI in both studies, strengthening the potential for these two genes to be promising genetic hits. The other gene that appeared across two separate studies was SLC6A3 (dopamine transporter gene). In addition to the modest association with PPI in the Greenwood et al. (2012) study, a genome-wide linkage analysis of the COGS sample suggested linkage (LOD score >2.2) for PPI on chromosome 5p15, a “gene dense” region that contains SLC6A3 (Greenwood et al., 2013), indicating that the dopamine transporter may be an additional gene of interest for follow up studies. Whether this endophenotype approach is more useful than genetic studies based on disease diagnosis is heavily debated in psychiatric genetics, but it is certainly complementary to GWAS studies of disease and may offer useful information regarding biological processes that cut across psychiatric diagnoses (Cuthbert and Insel, 2013).
2.3. Mutant Mouse Models: Where to go from here?
McCarroll et al. 2014 argue that a new “biological playbook” needs to be written to address the new genetic discoveries emerging from unbiased genome-wide studies (McCarroll et al., 2014). The question for molecular biologists and basic neuroscientists is - what potential genetic “hits” from association studies are plausible targets for follow-up biological studies? Since the genes identified in the PGC study are common variants with small effect, biological models would likely need to manipulate multiple genes to see a biologically relevant effect (Need and Goldstein, 2014). Biological interrogation of the genetic regions identified through GWAS are hindered by: (1) lack of clear functional effects of the identified SNPs, (2) the likelihood that multiple genes interact to produce the full manifestation of disease, (3) the identified risk alleles can be distal to the causative gene, and (4) the likely possibility that common variants modify disease risk produced primarily by rare variants (Arguello and Gogos, 2011). Thus, the ability to target specific genes or multiple genes in rodent models becomes daunting. Need & Goldstein (2014) suggest that a better approach may be to focus basic biological and model organism studies on more highly penetrant rare mutations such as chromosomal deletions or duplications identified by CNV analyses (Need and Goldstein, 2014), while continuing to interrogate the function of the 108 identified loci from the PGC study.
3. Neurodevelopmental models of schizophrenia
3.1. Neurodevelopmental risk factors
There is increasing evidence that schizophrenia has its roots in disrupted brain development due to both genetic and environmental risk factors, leading to psychosis emergence in adolescence and early adulthood (Cannon et al., 2003; Murray et al., 2002; Rapoport et al., 2012). Environmental risk factors are evident throughout development and include prenatal and perinatal risk factors, psychological risk factors in early life and adolescence, and exposure to drugs of abuse or trauma in adulthood. Several general factors such as season of birth (late winter/early spring) (Boyd et al. 1986; Machon et al. 1983; Mino & Oshima 2006; Torrey et al. 1997) and social factors such as urbanicity, immigrant status, and social isolation are associated with increased schizophrenia risk (Cannon et al., 2008; Dean et al., 2003; Marcelis et al., 1998). More specific risk factors include prenatal exposure to inflammation or birth complications, as well as adolescent exposure to drugs of abuse. Prior to discussing studies evaluating these risk factors in the context of gene × environment or environment × environment interactions, we first briefly review the evidence for the associated risk with schizophrenia. We focus on risk factors for schizophrenia because epidemiological studies of disease risk are what have produced candidate risk factors in model organisms in which PPI was measured.
3.2. Prenatal, perinatal, and early postnatal risk factors
Early life exposures to adverse environmental factors, either in utero or during the perinatal period, increase the risk of schizophrenia and include maternal stress, maternal malnutrition, immune activation or infections, or obstetric complications (Lewis and Levitt, 2002). PPI is a behavioral measure that has been extensively studied in many of these neurodevelopmental models as we have previously reviewed (Powell, 2010).
Epidemiological studies suggest an increased incidence of schizophrenia after exposure to viral or bacterial infections during early to mid gestation (reviewed in Brown and Susser, 2002; Fatemi and Folsom, 2009; Patterson, 2009; but see also Selten et al., 1999), with links being found between influenza (Mednick et al., 1988; O’Callaghan et al., 1991), bacterial infections (Sorensen et al., 2009), and also toxoplasmosis (Brown et al., 2005). These epidemiological studies have been supported by serological evidence of increased levels of gestational influenza infection (Brown et al., 2004a) and 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 individuals with schizophrenia. Animal studies have investigated the effects of maternal challenges with viral infection (e.g. influenza virus (Shi et al., 2003), immune activating agents such as the viral mimic polyriboinosinic-polyribocytidilic acid (PolyI:C), and bacterial endotoxin lipopolysaccharide (LPS) (for more thorough reviews see Estes and McAllister, 2016; Meyer, 2014; Meyer and Feldon, 2009a; Meyer and Feldon, 2009b; Patterson, 2009; Powell, 2010).
Prenatal nutritional deficiency has also been shown to increases the risk of schizophrenia (Brown and Susser, 2008; Susser et al., 1996; Xu et al., 2009). Thus, nutritional deficiency has been modeled in rodents by examining prenatal protein deprivation, which produces PPI deficits in offspring (Palmer et al., 2004). Maternal vitamin D deficiency also results in brain and behavioral abnormalities related to schizophrenia (reviewed in Burne et al., 2004a; Burne et al., 2004b; Burne et al., 2006; Eyles et al., 2013; Eyles et al., 2009; Kesby et al., 2006; Schoenrock and Tarantino, 2016) with some evidence of PPI deficits associated with vitamin D deficiency in rodents (Burne et al., 2004b; Kesby et al., 2006). Additionally, obstetric complications such as pre-eclampsia, cesarian section, and perinatal hypoxia have been well documented and linked to schizophrenia in several independent studies (Cannon et al., 2002; Hultman et al., 1997; Zornberg et al., 2000), and modeled in animals (reviewed in Boksa, 2004; Meyer and Feldon, 2009a; Powell, 2010).
There is an increased appreciation for the role of psychological stress, both prenatal and early and childhood, in the pathogenesis of schizophrenia (reviewed in Koenig, 2006; Koenig et al., 2002). The effects of prenatal stress on schizophrenia-related behaviors in animals have been mixed and depend on the methods of inducing “stress” in the pregnant dam (Koenig, 2006; Koenig et al., 2005; Lee et al., 2007; Lehmann et al., 2000). Childhood trauma or negative childhood experiences contribute to the development of neuropsychiatric disorders (Read and Bentall, 2012). The evidence for an association between psychosocial stress and psychosis is mixed with some studies showing an association between adverse life events and psychosis (Johns et al., 2004; Miller et al., 2001; Shevlin et al., 2008; Wiles et al., 2006), and other studies failing to see an association between adverse lifetime events and psychosis in high risk individuals (Cannon et al., 2016; Mason et al., 2004), or higher rates of childhood trauma in schizophrenia patients compared to controls (Kilian et al., 2017). Nevertheless, early postnatal stress has been assessed for its effects on schizophrenia-related behaviors with studies of more severe maternal deprivation (e.g. 24 hours) producing significant (Ellenbroek and Cools, 2000; Ellenbroek et al., 1998) or only mild or negligible deficits in PPI (Choy et al., 2009; Choy and van den Buuse, 2008), and shorter periods of maternal separation (e.g. 1–4 h/day) producing mild effects (Klug and van den Buuse, 2012) or no effect on PPI in rats (Finamore and Port, 2000; Weiss et al., 2001) or mice (Millstein et al., 2006). Many stress models, such as chronic unpredictable stress, social defeat stress, restraint stress, do not appear to affect PPI on their own. However, many of these manipulations have been used in combination with genetic risk factors (section 4.1) or other environmental manipulations (section 4.2) to affect PPI.
3.3 Adolescent Risk factors: social isolation and drugs of abuse
The juvenile/adolescent period is the time in which complex social behaviors develop and a critical period for remodeling of neural circuits important for social, emotional, and cognitive development (Casey et al., 2008; Giedd, 2008; Leon-Carrion et al., 2004). In schizophrenia, social withdrawal occurs early in the course of illness, prior to symptoms of psychosis, and predicts conversion to psychosis (Addington et al., 2008; Cannon et al., 2008; Moller and Husby, 2000). We have argued that social isolation and withdrawal in the course of schizophrenia can both trigger chronic stress cascades and be a consequence of the functional impairment resulting from premorbid social cognitive deficits in mental illness (Powell and Swerdlow, 2015). Because of the profound impact of social isolation and withdrawal on psychiatric health and the importance of the juvenile/adolescent period in social interaction and social development, post-weaning social isolation has been studied extensively in rodents. We refer the reader to our recent, more extensive review on the topic (Powell and Swerdlow, 2015).
Several studies have shown that drug abuse in adolescence increases the risk of developing schizophrenia (Nielsen et al., 2017), particularly adolescent cannabis use (Andréasson et al., 1987; Arseneault et al., 2002; van Os et al., 2002). Epidemiological findings suggest a link between cannabis use and psychosis (Gage et al., 2016; Vaucher et al., 2017) and that use of cannabis leads to an onset of psychosis at an earlier age than those who develop psychosis without a history of cannabis use (Barnes et al., 2006; Donoghue et al., 2014). However, the role of cannabis in schizophrenia risk is still unclear, and likely involves an increased susceptibility in genetically or environmentally susceptible individuals (Caspi et al., 2005; Di Forti et al., 2012; van Os et al., 2002). The effects of cannabis use on PPI are mixed (see Discussion for more detail).
4. “Two Hit” Models of Risk factors of Schizophrenia
As reviewed above, early developmental factors are implicated in the pathogenesis of schizophrenia (Davis et al., 2016), and recent GWAS have identified multiple common schizophrenia risk alleles contributing small effect to disease risk (Owen et al., 2016). In addition to common variants with small effects, there is also evidence for the involvement of several large CNVs in schizophrenia (Ross et al., 2006). Additionally, there are several non-genetic second-hits (substance abuse (McKetin et al., 2013), adolescent cannabis exposure (Moore et al., 2007), childhood abuse (Mortensen et al., 1999), and residential status (e.g. urbanicity; (Kelly et al., 2010) that act at different periods of neurodevelopmental stages to increase risk. Such factors might have relatively weak effects on their own but when acting at specific developmental stages in genetically susceptible individuals (Gene × Environment; G × E) or in individuals exposed to other environmental risk factors (Environment × Environment; E × E), these factor may lead to the development of schizophrenia. Evaluating causation of risk factors in human studies is difficult since the risk factors cannot be manipulated. One of the primary issues that is difficult to disentangle in human studies is whether risk factors are causal to disease or whether some other factor (or covariate) influences both the risk factor and disease (Kendler and Gardner, 2010).
During recent years much research has focused on “two hit” developmental animal models to fully understand the changes in brain anatomy and behavior present in schizophrenia. The timing of these hits during neurodevelopment is very important because they can cause differing outcomes, where early developmental hits can lead to more widespread abnormalities and later, or second hits, can cause more specific changes (Davis et al., 2016; Pantelis et al., 2003). Thus, many rodent studies have employed a “two hit” approach to test the hypothesis that maldevelopment during two critical time periods, e.g. early brain development and then adolescence, may lead to schizophrenia (Keshavan and Hogarty, 1999). Here we review recent research modeling combined genetic and environmental risk factors (G × E interactions) or combined environmental risk factors (E × E interactions) in preclinical studies with an emphasis on sensorimotor gating effects in the models.
4.1 Modeling Gene × Environment risk factors relevant to schizophrenia
Interaction between genetic risk factors and environmental stressors at specific developmental stages increases the chance of developing schizophrenia (Uher, 2014). Although the notion that schizophrenia results from a genetic predisposition followed by an environmental “hit” has been hypothesized for a number of years, only recently have these GxE interactions been evaluated in clinical studies (van Winkel et al., 2008). In human studies, GxE interactions have been reported for several candidate genes (Uher, 2014). One of the first examples showing an association between psychosis and a gene-environment combination was for the functional Val158Met polymorphism in catechol-O-methyltransferase (COMT). Specifically, individuals with the Val allele (i.e. the more efficient allele) that had used cannabis in adolescence had an increased risk for psychosis (Caspi et al., 2005); however, this initial study was not replicated in subsequent studies (De Sousa et al., 2013; Kantrowitz et al., 2009; Zammit et al., 2011; Zammit et al., 2007). Additionally, childhood maltreatment may interact with cannabis exposure and COMT to increase psychosis risk. Indeed, there have been two reports of an association between COMT genotype, childhood maltreatment, and cannabis (Alemany et al., 2014; Vinkers et al., 2013). Another gene-environment interaction was reported for the AKT1 gene and cannabis, with individuals carrying a polymorphism in AKT1 more likely to develop psychosis after cannabis use (Di Forti et al., 2012; van Winkel, 2011). Childhood maltreatment has been shown to interact with several genes to increase risk of psychosis, including BDNF (Alemany et al., 2011; although see Ramsay et al., 2013), FKBP5, a co-chaperone of the glucocorticoid receptor (Collip et al., 2013), and SLC6A4, encoding the serotonin transporter, (Aas et al., 2012). In utero infections have also been evaluated in the context of gene-environment interactions and psychosis risk. For example, GRIN2B, a component of NMDA glutamate receptors, interacted with exposure to herpes simplex virus-2 in utero (Demontis et al., 2011), and preliminary evidence from systematic gene-environment interaction studies indicated that CTNNA3, which encodes a cadherin-associated protein, interacted with in utero cytomegalovirus exposure in schizophrenia cases (Borglum et al., 2014). Thus, there is increasing evidence for gene-environment interactions in schizophrenia risk. Whether or not these gene-environment interactions are relevant to sensorimotor gating in humans has yet to be determined. Nevertheless, some of these gene-environment interactions have been studied in animal models and here we summarize how GxE interactions have affected PPI in animal studies (Table 1).
Table 1.
Susceptibility Gene | Model description/background/rationale | Second Hit | Effects on PPI | Prevention/Treatment strategies | References |
---|---|---|---|---|---|
DISC1 | |||||
DN-DISC1(dominant-negative DISC1 under the expression control of CaMKII promoter (DN-DISC1; line 10) | DISC1 plays important role in brain development | PolyI:C 5 mg/kg or saline (PND2-6) | Ø PPI & Ø PA in WT vs TG, PolyI:C/WT vs | Ibi et al. 2010 | |
DISC1-L100P | Missense mutation in exon 2 of DISC1 at L100P in mice | Prenatal PolyI:C (2.5 mg/kg) on GD 9 | Ø PPI in poly I:C vs Sal, ↓PPI in DISC1-L100P & DISC1-L100P/Poly I:C vs WT & L100P/Sal | Co-administration of IL-6 antagonist PPI deficits in DISC1-L100P/poly I:C mice | Lipina et al 2013. |
DISC1-Q31L | Prenatal PolyI:C (5 mg/kg) on GD | 16wks: ↓PPI in poly I:C vs Sal, ↓PPI in DISC1-Q31L vs WT. No potentiatio n of PPI deficit in DISC1-Q31L/Poly I:C | Lipina et al 2013 | ||
DISC1 (Transgenic model of inducible expression of dominant negative human hDISC1 in forebrain) |
Prenatal exposure to Pb++ | Ø PPI in Pb++ vs Reg ↓PPI in F hDISC1/Pb++ vs hDISC1/Reg mice. | Administration of D-serine, reversed PPI hDISC1/Pb++ in F | Abazyan et al. 2014 | |
DISC1 DISC1 (L10P and Q31L mutants) |
Chronic social defeat stress | Ø PPI in WT/NS vs WT/CSD & Q31L+/−/NS vs Q31L+/−/CSD; ↓PPI in L100P+/−/NS vs WT/NS; No GxE | Haque et al,2012 | ||
Nurr1 | |||||
Nurr1 (Heterozygous constitutive deletion of Nurr1) |
Nurr1 plays important role in differentiation, migration and survival of DA ergic neurons | Prenatal PolyI:C (5mg/kg) on GD9 | ↓PPI in Nurr1 +/− vs WT. ↓PPI in WT/poly I:C vs WT/Veh; ↓PPI in Nurr1 +/−/PolyI:C mice | Vuillermot et al., 2012 | |
Nurr1 (Nurr1-null mice by homologous recombination at exon 3) |
Social Isolation | Ø PPI in WT/Iso vs WT/Soc. ↑PPI in Nurr1+/−/Soc vs WT/Soc. ↓PPI in Nurr1+/−/Iso vs WT/Iso and WT/Soc. | Eells et al 2006 | ||
Nurr1 | Infection with Toxoplasma gondi | ↓Baseline startle & Ø PPI in Nurr1+/− vs WT in M&F. Ø PPI after infection | Eells et al 2015 | ||
Neuregulin | |||||
NRG1 TM HET (HET NRG1 transmembrane domain knock-out targeting exon 11) |
NRG1 may represent a SZ susceptibility gene. Involved in neuronal migration, synaptogenesis, neuronglia interactions. | Acute and subchronic administratio n of CP55,940 (0.4mg/kg) | ↓PPI in NRG1 HET/Veh vs WT/Veh. ↓PPI in WT/CP at 74dB. ↑PPI in HET/CP vsWT/CP | Boucher et al 2011 | |
NRG1 TM HET | THC (5 & 10mg/kg) | Ø PPI in WT mice, THC ↑PPI in NRG1 +/− mice; ↓PA in NRG1 +/− at high dose (10 mg/kg) | Boucher et al 2007 | ||
NRG1 TM HET | Prenatal poly I:C on GD 9 (5mg/kg). Pups were crossfostered from PND 0–2 | ↓PPI in NRG1 +/−, NRG1 +/−/PolyI:C & ↓PPI inNRG1+/−poly I:C infected dams and cross-fostered to poly I:C infected dams | O’Leary et al 2014 | ||
NRG1 TM HET | THC (10 mg/kg, IP) 21 days during adolescence (PND31–52) | Ø PPI after acute or chronic THC or following washout in WT or Het mice; Ø PPI in NRG1 Het | Long et al. 2013 | ||
Reelin | |||||
Reelin KO | Reelin levels in brains of SZ are reduced. | Prenatal Hypoxia | ↓PPI by hypoxia in both WT and HET mice. No GxE interaction | Howell & Pillai 2016 | |
Reelin HRM | CORT treatment | ↓PPI in WT CORT vs WT CON in M not in F, Ø PPI in HRM CON vs WT CON & HRM/CORT vs WT CORT | Schroeder et al. 2015 | ||
PACAP | |||||
PACAP KO, M | PACAP affects neurotransmission; potential SZ susceptibility gene | Social Isolation | ↓PPI in KO vs. WT (PA data not shown); ↓PPI in WT/Iso vs WT/Soc. ↓PPI KO/Iso vs WT/Iso. | EE: ↓PPI in KO & WT | Ishihama et al. 2010 |
SNAP25 | |||||
SNAP25 (Blind drunk mutant) |
Snap25 is a SNARE protein and is linked to SZ | Prenatal Stress | ↓PPI in WT vs Brd mutants in NS, PNS ↓PPI in WT vs Brd mutants. ↓PPI in PNS Brd mutants than NS mutants. Significant GxE interaction | CLO: Reversed PPI in NS & PNS Brd mutants | Oliver and Davies, 2009 |
BDNF | |||||
BDNF | BDNF is involved in brain development and neuroplasticity and is implicated in pathophysiology of SZ | Chronic METH during late adolescence and early adulthood | ↓PPI in HET M&F mice; Ø PPI in METH- treated mice; No GxE interaction on baseline PPI | Manning & van den Buuse, 2013 | |
BDNF | CP (0.4mg/kg) during young adulthood | Ø Baseline PPI following G or E insult. ↓PPI in HET vs WT. ↑PPI in HET/CP vs HET/Sal M after acute CP challenge. ↑PPI in WT/Veh, WT/CP, HET/Veh and HET/CP after acute CP challenge. |
Klug & van de Buuse 2013 | ||
Tap1 | |||||
Tap1 KO mice, | Prenatal influenza infection associated with increased SZ risk. Transporter associated with antigen processing 1 mice have reduced expression of MHC class I. | Neonatal influenza | Influenza infection on PND 3 or 4: ↓PPI in adult KO mice (during ISI block), trend for ↑PA in infected mice in startle threshold block; Ø PPI & Ø PA in infected vs. non-infected WT C57 mice | Asp et al. 2010 | |
GCPII | |||||
GCPII | GCPII Glutamate carboxypeptidase II catalyzes NAAG and is also involve in dietary folic acid metabolism |
Dietary folic acid deficiency | Ø PPI in WT FD diet vs WT control diet & GCPII Het vs WT & GCPII Het/FD diet vs WT control diet. | Schaevitz et al., 2012 | |
NMDA Receptor | |||||
NR1 (Ppp1r2-Cre/floxed-GluN1mice has GluN1 deletion in a subset of cortical interneurons) |
NR1 NMDA receptor hypofunction is implicated in schizophrenia. NR1 knock out mice have early postnatal deletion of NMDA receptor from corticolimbic interneurons |
Social isolation | ↓PPI in NR1 KO vs WT, Ø PPI in WT/SI vs WT/Soc. ↓PPI in NR1KO/SI & NR1 KO/Soc vs WT/Soc | Chronic apocynin (from postnatal week 2) prevented ↓PPI in NR1/Soc and NR1/SI | Jiang et al. 2013 |
Abbreviations: BDNF brain derived neurotropic factor, Brd blind drunk mutant, CaMKII Ca2+/calmodulin-dependent protein kinase II, CLO clozapine,CORT corticosterone, CP CP55,940, CSD chronic social defeat, dB decibel, DISC-1 Disrupted-In-Schizophrenia-1, DIZ dizocilpine, DN dominant-negative, EE environmental enrichment, F female, FD folate deficient, G gene, GCPII Glutamate carboxypeptidase II, GD gestational day, HRM heterozygous reelin mice, IL interleukin, ISI interstimulus interval, KO knockout, M male, METH metamphetamine, NAAG N-acetylalpha L-aspartyl-L-glutamate, NR1 NMDA receptor subunit, NRG neuregulin, ns not significant(ly), NS non stressed, Nurr1 nuclear receptor, PA magnitude of response to pulse alone, PACAP pituitary adenylate-cyclase-activating polypeptide, Pb++ lead, PND postnatal day, PNS prenatal stress, PolyI:C polyinosinic: polycytidylic acid, PP prepulse, PPI prepulse inhibition of startle, Reg regular diet, Sal saline, SCID Severe combined immunodeficiency, SI social isolation, SNAP-25 synaptosomal-associated protein of 25kDa, SNP single nucleotide polymorphism, SZ schizophrenia, TG transgenic, THC tetrahydrocannabinol, TM transmembrane domaine, WT wild-type, ↓ decreased, ↑ increased, Ø unchanged, −/− homozygous mice, +/− heterozygous mice
4.1.1. Disrupted-in-schizophrenia 1 (DISC1)
Disrupted-in-schizophrenia 1 (DISC1) was one of the first genes implicated in the pathophysiology of schizophrenia based on studies in a large Scottish family. The mutation involves a balanced chromosome translocation on chromosome 1q42 (Blackwood et al., 2001; Millar et al., 2000). DISC1 is a synaptic protein involved in cell proliferation, differentiation, and migration (Brandon and Sawa, 2011; Jaaro-Peled et al., 2009). Several different lines of transgenic mice containing DISC 1 gene mutations have been created to investigate its role in behavior and brain development (Ji et al., 2014). Here we summarize the current studies investigating DISC1 mutant mice combined with environmental risk factors.
DISC1 × maternal immune activation
Lipina and colleagues created two point mutation mouse lines DISC1-L100P and DISC1-Q31L and showed that the schizophrenia related phenotypic effects were more pronounced in DISC1-L100P mice (Lipina et al., 2012; Lipina et al., 2011). Combining this point mutation with maternal immune activation by administration of a sub-threshold dose of PolyI:C (2.5 mg/kg) at GD 9 led to more robust PPI deficits and decreased startle amplitude in DISC1-L100P offspring compared to wildtype control mice. When MIA was combined with the DISC1-Q31L mutation, both DISC1-Q31L and PolyI:C (5 mg/kg) produced PPI deficits at 16 weeks, but these deficits were not further potentiated by their combination. Following the gestational exposure of PolyI:C, increased levels of interleukin-6 (IL-6) were more pronounced in DISC1-L100P compared to DISC1-Q31L mice or wild type controls. When an IL-6 antagonist was co-administered at the time of maternal immune activation, PPI deficits were rescued in DISC1-L100P mice (Lipina et al., 2013).
DISC1 × neonatal immune activation
Ibi and colleagues utilized transgenic dominant-negative mutant DISC1 to study genetic and environmental risk factors by injecting poly I:C (5mg/kg) between PND 2 & 6. Neither DISC1 mutation nor neonatal poly I:C administration produced any changes in PPI (Ibi et al., 2010).
DISC1 × prenatal lead exposure
Recently, prenatal lead exposure has been associated with an increased susceptibility of schizophrenia in adulthood (Opler et al., 2004). Lead (Pb++) is a potent antagonist of NMDA receptors and it is possible that Pb++ contributes to schizophrenia in genetically vulnerable individuals. Hence, another study examined the interaction of inducible mutant human DISC1 (mhDISC1) with prenatal exposure to lead. Female, but not male, mhDISC1 mice with prenatal lead exposure showed mild PPI impairments (at low prepulse intensities 74 and 78dB) (Abazyan et al., 2014).
DISC1 × social defeat stress
Interactions between DISC1 point mutations and chronic social defeat have also been examined in DISC1-L100P and Q31L mice. Mutant DISC1-L100P mice showed lower PPI than WT mice and DISC1- Q31L mice. Chronic social defeat stress did not affect PPI in WT, DISC1-L100P, or Q31L mice. Thus, there was no evidence of significant gene × environment interactions for social defeat stress and DISC1 mutations on PPI (Haque et al., 2012).
4.1.2. Nuclear receptor related 1 protein
The nuclear receptor related 1 (Nurr1) protein is a member of the orphan steroid hormone receptor family. Nurr 1 is expressed in mesencephalic dopaminergic neurons and is critical for their survival and differentiation (Kadkhodaei et al., 2009; Rojas et al., 2007). Nurr1 heterozygous mice showed reduced dopamine in both the mesolimbic and mesocortical dopamine pathways, suggesting that Nurr1 is involved in the maintenance of dopamine neurotransmission (Eells et al., 2002).
Nurr1 × maternal immune activation
To investigate the interaction of maternal immune activation and Nurr1, Nurr1 mutant (heterozygous deletion of the Nurr1 gene) mice were exposed to PolyI:C on GD 9. When tested in adulthood (PND 75-120), the combination of Nurr1 mutation and PolyI:C resulted in additive effects on PPI, with both genotype and gestational exposure exerting main effects and Nurr1 (+/−) mice exposed to PolyI:C showing the most pronounced PPI deficits (Vuillermot et al., 2012).
Nurr1 × social isolation
Another study investigated the interaction between Nurr1 and early postnatal social isolation. In this study, WT and Nurr1 null heterozygous mice subjected to social isolation from weaning were tested for PPI alterations after 12 weeks of isolation. Nurr1 heterozygous miceshowed decreased PPI after social isolation, with normal PPI associated with isolation rearing or genotype alone (Eells et al., 2006). Thus, social isolation potentiated the effects of Nurr1 mutation on PPI.
Nurr1 × infection
In a recent study, Nurr 1 (+/−) mice were infected with Toxoplasma gondi and tested in a behavioral battery of tests relevant to schizophrenia. Nurr 1 (+/−) mice showed reduced startle magnitude but no differences in PPI compared to WT controls before the infection. There were no gene × environment interactions in startle magnitude or PPI in male or female mice when tested 6 weeks after the infection (Eells et al., 2015).
4.1.3. Neuregulin 1
Another susceptibility gene studied for its association to schizophrenia and sensorimotor gating is Neuregulin 1 (NRG1). Indeed, a schizophrenia-related NRG1 polymorphism has been associated with prepulse inhibition in human controls (Roussos et al., 2011) and schizophrenia patients (Greenwood et al., 2011). NRG1 is involved in neuronal migration, synaptogenesis, and neuron-glial interactions in the developing brain, as well as excitatory and inhibitory neurotransmission in the adult brain (Harrison and Law, 2006). Because of the role in schizophrenia and brain development, NRG1 is an intriguing target to examine gene-environment interactions.
NRG1 × cannabinoid administration
The effects of cannabinoids in NRG1 mutant mice have been examined in several studies with some limited evidence of differential sensitivity to the behavioral effects of tetrahydrocannabinol (THC). In one study THC increased PPI and reduced startle in NRG1 HET mice, but not in WT mice (Boucher et al., 2007). Chronic adolescent exposure to THC (10 mg/kg; 21 days) had no effect in either NRG1 HET or WT mice (Long et al., 2013). In a subsequent study, Boucher and colleagues (Boucher et al., 2011) investigated the interaction of NRG1 and cannabinoid administration during adulthood. Single administration of the synthetic cannabinoid CP55,940 decreased PPI in WT mice and increased PPI in NRG1 HET mice. On the other hand, CP55,940 decreased acoustic startle in both WT and NRG1 HET mice. Thus, there is limited evidence for an interaction between NRG1 and cannabinoids on PPI and startle, but the data do not suggest that NRG1 and cannabinoids have an additive or synergistic effect on reducing PPI; if anything, cannabinoids produced increases in PPI.
NRG1 × maternal immune activation
Another study explored the effects of maternal immune activation in heterozygous NRG1 TM-domain mice. PolyI:C- or saline-exposed offspring were further subjected to cross fostering. Although there was an overall effect of decreased PPI in NRG1 HET mice, no clear interaction between NRG1 genotype and PolyI:C treatment was observed. There was some evidence of a potentiation of the PPI deficit in NRG1 HET with PolyI:C exposure, but this interaction was not consistent and interacted in a complicated way with cross-fostering (O’Leary et al., 2014).
4.1.4. Other susceptibility genes
Reelin × hypoxia
Reelin glycoprotein is involved in synaptic plasticity and brain development. Reductions in Reelin mRNA and protein levels have been found in prefrontal cortex, hippocampus, and cerebellum of schizophrenia patients (Cassidy et al., 2010a; Cassidy et al., 2010b). To investigate gene-environment interactions Reeler mice (haploinsufficient for Reelin) were exposed to prenatal hypoxia (9% oxygen) on GD17 for 2 hours. PPI was assessed at 3 months of age in mice exposed to hypoxia (9% oxygen) and normoxia in utero. PPI was increased in both WT and Reeler mice prenatally exposed to hypoxia. Interestingly, startle amplitude was decreased in WT hypoxia mice and Reeler normoxia mice compared to control WT normoxia mice (Howell and Pillai, 2016). Thus, the combination of Reelin haploinsufficiency and hypoxia did not produce PPI deficits.
Reelin × corticosterone
To model HPA increases during chronic stress, male and female heterozygous Reeler mice received chronic corticosterone treatment in the drinking water for 21 days starting at 6 weeks of age. Corticosterone treatment reduced PPI in male WT mice but had no effect on PPI in male reeler mice, suggesting a potential protective effect of reelin deficiency (Schroeder et al., 2015).
PACAP × social isolation
Pituitary adenylate cyclase-activating peptide (PACAP) is a member of the vasoactive intestinal peptide (VIP)/secretin/glucagon superfamily and is distributed widely in both brain and periphery (Waschek, 2013). Mice lacking Adcyap1 gene encoding PACAP (−/−) display several schizophrenia-related behavioral phenotypes that can be reversed by antipsychotics (Hashimoto et al., 2009; Hashimoto et al., 2001; Hashimoto et al., 2007; Tanaka et al., 2006). Four week old PACAP-null mutant and WT mice were housed in social isolation for two weeks. PACAP KO group-reared mice showed decreased PPI compared to WT group-reared mice. Social isolation also disrupted PPI in WT mice. PACAP KO mice reared in social isolation showed profound decrease in PPI compared to WT group-reared mice, indicating a significant G × E interaction in this model (Ishihama et al., 2010).
SNAP25 × prenatal stress
Synaptosomal-associated protein-25 (SNAP25) is a SNARE protein known to play a role in neurotransmitter release (Chen and Scheller, 2001) and long-term potentiation (Jurado et al., 2013). GWAS have indicated the involvement of the SNAP-25 genomic region in schizophrenia (Lewis et al., 2003). Additionally, altered levels of SNAP-25 were found in frontal cortex (Honer et al., 2002) and hippocampus (Davidsson et al., 1999) of patients with schizophrenia. Based on this genetic association, Jeans et al. conducted a GxE study utilizing blind-drunk (Bdr) mice that express defective SNAP25 protein due to a single amino acid substitution (167T) that disrupts the normal recycling of synaptic vesicles (Jeans et al., 2007). In this study Bdr mice were exposed to repeated prenatal variable stress carried out from GD 11.5 to 17.5. Male offspring were assessed in a battery of behavioral tasks at the age of 8–11 weeks. Brd mutants showed a significant PPI deficit compared to WT controls. Brd mutants exposed to prenatal stress showed enhancement of PPI deficits compared to non-stressed Brd mutants and prenatally stressed controls. These deficits were ameliorated by administration of clozapine in Brd mutants from both the stressed and non-stressed groups (Oliver and Davies, 2009), suggesting some degree of predictive validity in the model.
BDNF deficiency × adolescent drug abuse
Brain-derived neurotropic factor (BDNF) is growth factor involved in brain development and neuroplasticity, and altered BDNF signaling is reported in schizophrenia (Autry and Monteggia, 2012). Thus, a recent study evaluated the effects of repeated synthetic cannabinoid CP55,940 exposure for 3 weeks starting at PND42 in BDNF deficient mice. BDNF HET mice showed decreased PPI compared to WT mice, and repeated administration of CP55,940 did not alter PPI in WT females or males. Acute challenge with CP55,940 increased PPI, particularly in the “double hit” group (BDNF HET male mice exposed to chronic CP55,940) but these effects should be interpreted with caution since acute CP55,940 reduced startle magnitude, which could confound PPI results (Klug and van den Buuse, 2013). Thus, chronic CP55,940 did not produce the expected potentiation of PPI deficits in BDNF HET mice. Chronic adolescent methamphetamine administration was also examined for its effects in BDNF HET mice. While both male and female BDNF HETs showed decreased PPI, chronic METH during adolescence did not potentiate these PPI deficits or have an effect on its own (Manning and van den Buuse, 2013).
Tap 1 knock out × neonatal influenza A virus
Another study that looked at G × E interaction infected immunodeficient Tap 1 mice (due to targeted disruption of the gene encoding MHC class 1 and therefore lack functional CD8+ T cells) with influenza virus during the neonatal period (PND 3&4). Virus-infected Tap 1 KO mice, but not WT control mice, had impaired PPI, indicating long-term deficits in sensorimotor gating in immune deficient mice exposed to neonatal infection (Asp et al., 2010).
GCPII × folate deficiency
Glutamate carboxypeptidase II is a neuropeptidase that is present in astrocytes and catalyzes N-acetylaspartylglutamate (NAAG) into glutamate and N-acetylaspartate (Berger et al., 1999; Luthi-Carter et al., 1998). GCPII is also involved in dietry folate metabolism and absorption (Devlin et al., 2000), and thus the combined effect of dietry folic acid deficiency and mutation of GCPII was examined in mice. On PND25 GCPII (heterozygous mice) were either assigned to control folate diet (2 mg/kg) or folate deficient diet (0.3 mg/kg folate). The combination of GCPII mutation and folic acid deficiency did not affect PPI (Schaevitz et al., 2012).
NMDA receptor × social isolation
Jiang et al, 2013 examined G × E interaction by postnatal deletion of NMDA receptor in subset of cortical interneurons combined with post-weaning social isolation in mice (Jiang et al., 2013). In this study Ppp1r2-Cre/floxed-GluN1 (NR1 KO) mice were generated that have 40–50% deletion of NR1 in inhibitory interneurons by PND21. NR1 KO mice showed impaired PPI compared to WT controls. Isolation rearing alone did not produce PPI deficits; however, PPI was impaired in NR1 KO mice reared in social isolation. Chronic treatment with apocynin (starting at the age of 2 weeks) prevented PPI deficits in NR1 KO group-reared and isolation-reared mice (Jiang et al., 2013).
4.2 Environment × Environment interactions
Many environmental risk factors have been implicated in schizophrenia such as malnutrition, prenatal exposure to infections, stress during neonatal and postnatal development, and substance abuse (reviewed in Section 3). There is evidence of additive effects of urbanicity, cannabis use, and childhood trauma on risk of psychotic experiences (Guloksuz et al., 2015) or between cannabis use and childhood trauma (Harley et al., 2010; Houston et al., 2008; Houston et al., 2011; Konings et al., 2012; Murphy et al., 2013). This section focuses on the role of two environmental “hits”, or environment-environment interactions, in rodent models of PPI.
4.2.1. Immune dysregulation or Infections
It is well established that in utero or maternal exposure to infection is associated with increased risk of schizophrenia in offspring (Brown and Susser, 2002). The emergence of schizophrenia pathology in infected offspring during adulthood depends on the timing of infection during gestational period (Buka et al., 2008; Cheslack-Postava et al., 2015). Of course, all individuals who are exposed to infections in utero do not go on to develop schizophrenia, thus consideration of genetic susceptibility and/or additional environmental insults are important. Animal models of maternal infection include gestational exposure to either the TLR 3 agonist polyinosinic-polyctidylic acid (poly I:C), a synthetic analogue of double-stranded RNA, or lipopolysaccharide (LPS), a bacterial endotoxin that activates TLR 4.
Maternal immune activation × stress
Several recent studies have evaluated the interaction of maternal immune activation and juvenile/adolescent stress in mice. Giovanoli et al. (2016) exposed pregnant dams to a subthreshold dose of PolyI:C (1 mg/kg) or saline on GD 9 and offspring were subsequently exposed to varied unpredictable stress between postnatal day 30 and 40 (i.e. Electric foot shock, restraint stress, swimming stress, food deprivation, repeated home cage changes applied to alternate days). Neither prenatal immune activation nor stress alone affected PPI; however, peripubertal unpredictable stress was associated with disruption of PPI in offspring born from Poly I:C-infected mice but not saline controls. Thus, the combination of maternal immune activation and peripubertal stress disrupted PPI. Preventive treatment with minocycline (tetracycline antibiotic) before and throughout the exposure of stress prevented the PPI deficits in stressed PolyI:C offspring (Giovanoli et al., 2016).
An earlier study investigated the combined effects of maternal immune activation on GD 12 (20mg/kg) and juvenile stress in C57BL/6 mice. Pups born form poly I:C-infected dams were subjected to restraint stress for 3 consecutive days from PND 33 to 35. Juvenile restraint stress or gestational Poly I:C alone did not alter PPI when tested 24 hours after the last stress episode on PND 36; however, mice exposed to both maternal immune activation and juvenile restraint stress did show PPI deficits (Deslauriers et al., 2013). Administration of the antioxidant α-lipoic acid before restraint stress prevented the PPI deficits in the two-hit group and reduced oxidative stress levels in frontal cortex (Deslauriers et al., 2014). Other studies have failed to show a potentiating effect of stress on maternal immune activation-induced PPI deficits in rats (Yee et al., 2011), suggesting potential species differences in the interaction. Nevertheless, the prevention of some of the behavioral effects of the combined maternal immune activation and juvenile/pubertal stress with drugs targeting inflammation or oxidative stress indicates that this combined model may be useful in drug development.
4.2.2. Social isolation
As reviewed above, social isolation in rodents is a developmental manipulation in which rodents are raised singly-housed in absence of any social interaction with other rats or mice. In this section we summarize the effects of combining social isolation with other developmental insults on PPI.
Neonatal domoic acid × social isolation
A recent study investigated the interaction between neonatal domoic acid injections and isolation rearing in Sprague-Dawley rats. Domoic acid is an AMPA/kainite agonist that, when administered during the second postnatal week, results in later onset of behavioral phenotypes consistent with schizophrenia (Burt et al., 2008a, b). In this dual-hit study, pups were injected with domoic acid (20 μg/kg s.c.) from postnatal day 8–14 and then assigned to group-housing or social isolation at weaning. Isolated rats showed PPI deficits. Interestingly, domoic acid treatment increased PPI in isolates but no effects were found in group housed animals (Marriott et al., 2016).
Social isolation × methamphetamine
Another study examined E × E interaction by utilizing social isolation from weaning and chronic methamphetamine administration. Female Wistar rats were reared in social isolation or group housing from weaning. Another environmental hit was added by administering escalating doses of methamphetamine (2–6 mg/kg b.i.d) for 16 days from PND 35 to 50. On PND 78, female rats were tested in PPI. Social isolation alone reduced PPI in rats, and chronic administration of methamphetamine reduced PPI to the same extent in isolation-reared and group-housed controls (Strauss et al., 2014).
Neonatal NMDA antagonist × Social isolation
Lim and colleagues examined E × E interaction by combining perinatal MK801 treatment with social isolation in Sprague-Dawley rats. Rats were injected with MK801 (0.2 mg/kg) from PND 7 to 10 and either isolated or group housed at the time of weaning (PND 21). When tested in adulthood (PND 91) rats exposed to MK-801 and social isolation showed robust PPI deficits (Lim et al., 2012). Similarly, Gaskin et al evaluated the effects of two developmental insults by combining neonatal phencyclidine (PCP) injections and social isolation from weaning in Lister-hooded rats. Rats that received both insults showed deficits in PPI, which were not present in groups subjected to social isolation or neonatal PCP administration alone (Gaskin et al., 2014). These studies indicate that neonatal NMDA antagonist administration, combined with post-weaning social isolation, produce robust disruptions in PPI.
4.2.3. Maternal Separation
Maternal separation × Conditioned avoidance × PCP
Another study utilized a multiple hit approach during different developmental stages in Sprague- Dawley rats to examine combined environmental insults (Chen et al., 2011). Rats were subjected to maternal separation from PND 3 to 10 (first-hit) and then to avoidance conditioning on PND 49–56 (second-hit) and injected with PCP (3 mg/kg) immediately after each avoidance training (third-hit). The three hits were then assessed for their effect on change in %PPI from adolescence to adulthood. Maternal separation blocked the adolescent to adult increase in PPI observed in saline-treated rats but this effect was not evident in rats exposed to avoidance training, suggesting that the second hit remediated some of the effects of the first hit. This study demonstrates the complexity of using multiple “hits” within the same experiment, when triple interactions are being tested.
Maternal separation × corticosterone treatment
In this study Wistar rats underwent maternal deprivation on PND 9 for 24 h and then corticosterone treatment for 2 weeks in young adulthood (starting at 8 weeks of age). There was no effect of maternal separation, chronic corticosterone, or their combination on baseline PPI. Apomorphine disrupted PPI in all groups except those sustaining the combination of maternal separation and chronic corticosterone; whereas, amphetamine disrupted PPI in all groups except the maternally deprived groups. Thus, rather than an increased sensitivity, rats exposed to maternal deprivation and early adult corticosterone showed a decreased sensitivity to dopamine agonists (Choy and van den Buuse, 2008).
5. Discussion
Here we summarize schizophrenia risk factors, neurodevelopmental animal models, and the current findings from two hit models of these risk factors published in recent years. As reviewed above, we focused on PPI because of its strong relationship with schizophrenia, its heritability, and its sensitivity to developmental risk factors. Taken together, the studies suggest that some gene and environment combinations result in more pronounced PPI deficits than either manipulation alone. For example, post-weaning social isolation potentiates the PPI deficits in inhibitory neuron-specific NR1 KO mice (Jiang et al., 2013), PACAP KO mice (Ishihama et al 2010), and Nurr1 HET mice (Eells et al., 2006), and prenatal stress potentiates PPI effects in SNAP25 (Brd) mutants (Oliver and Davies, 2009). Inflammation is another second hit that has been shown to enhance PPI deficits in genetic mutants. Neonatal influenza produced PPI deficits in immunodeficient Tap 1 KO mice (Asp et al., 2010), and maternal immune activation with PolyI:C increases PPI deficits in Nurr1 HET mice (Vuillermot et al., 2012) and DISC1 mutant mice (Lipina et al., 2013) but failed to interact with NRG1 HETs (O’Leary et al., 2014). The evidence for adolescent cannabinoids interacting with genetic risk factors is less compelling. There was no evidence that THC or the synthetic cannabinoid agonist CP55,940 potentiated the effects of genetic risk factors on PPI (Long et al., 2013). In fact, where there was an interaction with genotype, cannabinoids actually increased PPI in the mutant mice (Boucher et al., 2007; Boucher et al., 2011; Klug and van den Buuse, 2013). Considering that the effects of cannabis use on PPI are mixed in the clinical literature, these results may not be surprising. In adults, chronic marijuana use was not associated with PPI (Quednow et al., 2004); however, adult marijuana users that initiated use during adolescence had decreased PPI compared to non-using controls in a task that involved attending to the auditory stimuli (Kedzior and Martin-Iverson, 2006, 2007; Scholes and Martin-Iverson, 2009). The timing of marijuana initiation may be important to the effects on PPI. In fact, there is evidence that cannabis use increased PPI in “at risk” and “early psychosis” subjects (Cadenhead, 2011). Human studies indicate that age of cannabis use onset, duration of use, and stage of illness at the time of PPI testing contribute to the effects of cannabis use on PPI. Thus, recapitulating these effects in animal models may be particularly challenging, and it is not clear the direction of prediction (i.e. increased or decreased PPI) considering the equivocal effects of cannabis use on PPI in humans. Animal studies of cannabinoids and PPI are equally equivocal. Acute and repeated juvenile and peri-pubertal administration of the cannabinoid agonist, WIN55,212-2, disrupted PPI in adulthood (Schneider et al., 2005; Schneider and Koch, 2002, 2003; Wegener and Koch, 2009); however, other groups have failed to replicate these findings (Bortolato et al., 2005; Bortolato et al., 2014). Adolescent exposure to other drugs of abuse, including amphetamine and alcohol, do not appear to affect PPI in adulthood (Coleman Jr et al., 2011; Richetto et al., 2013). Thus, there is not a lot of compelling evidence from animal studies that exposure to drugs of abuse in adolescence has an enduring effect on PPI. It should be noted, however, that animal studies use either THC or synthetic cannabinoid agonists; whereas, humans smoke cannabis, which contains many constituents in addition to THC, making it difficult to model drug exposure in model organisms. These clinical and preclinical studies of cannabis and PPI suggest that PPI may not be the most relevant measure of the link between cannabis use and psychosis.
Regarding double hits of two environmental/developmental risk factors, there is evidence for the combined effects of maternal immune activation and adolescent stress on PPI in mice (Deslauriers et al., 2013; Giovanoli et al., 2013), but not in rats (Yee et al., 2011). Additionally, perinatal NMDA antagonism combined with post-weaning social isolation produced deficits in PPI in rats (Gaskin et al., 2014; Lim et al., 2012). Many of these psychosocial stressors and neonatal/prenatal immune activation manipulations, as well as risk gene models, often have effects on their own, making it difficult to determine additivity or synergy in the combined models or producing ceiling effects in which further disruption is not achievable. For example, although both social isolation and adolescent methamphetamine reduced PPI in rats, the combination had no additive or synergistic effect (Strauss et al., 2014). Similarly, prenatal hypoxia produced PPI deficits in Het and KO Reeler mice (Howell and Pillai, 2016). To address the issue of main effects and to provide adequate behavioral windows to assess potentiation, many studies have used sub-threshold manipulations (e.g. lower, sub-threshold dose of PolyI:C as used in Giovanoli et al., 2013) and/or heterozygous mutant mice to assess effects of double hits. This approach may provide models that better mimic the nature of genetic and environmental interactions in the human population.
Most of the G × E models we reviewed looked at one susceptibility gene and environmental factors; however, schizophrenia involves more than one gene (Owen et al., 2016) and thus studies will likely also begin examining multiple risk genes to investigate epistatic interactions. Additionally, most of the studies reviewed focused on the neuronal function of susceptibility genes; however, the function of these genes in other cells such as glia should also be considered. For example, DISC1 is expressed in astrocytes and microglia (Seshadri et al., 2010). Mutant DISC1 expressed in astrocytes decreased production of D-serine in astrocytes, which was associated with a greater response to MK-801 in PPI (Ma et al., 2013). Similarly, astrocytes also produce BDNF (Girardet et al., 2013; Sun et al., 2014) and overexpression of BDNF in hippocampal astrocytes produces anxiolytic and antidepressant-like effects (Quesseveur et al., 2013); however, behavioral changes relevant to schizophrenia have not been evaluated. It will be interesting to look at the role of these susceptibility genes or multiple developmental insults in microglia and/or astrocytes in GxE or ExE interactions.
One observation emerging from these studies is sex-specific effects of G × E and E × E interactions. In schizophrenia, sex-specific effects are observed in the course and symptoms of the illness with males having an earlier onset than females, thus it is not surprising that animal models related to the disease report different effects in males and females. Additionally, schizophrenia is increasingly considered a neurodevelopmental disorder (Section 3.0) and the developmental timing of environmental insults can greatly impact the pattern of results in the model. For example, maternal immune activation by administration of Poly I:C at early and late gestation affect behavior of the offspring differently (Meyer et al., 2006; Smith et al., 2007). Similarly, the effects of postnatal hypoxia on PPI depend on the timing and severity of the hypoxia. For example, hypoxia at PND 9 had no effect on PPI even though it altered mesolimbic dopamine neurochemistry (Sandager-Nielsen et al., 2004). Sub-chronic exposure to hypoxia from PND 4–8 did produce PPI deficits in adult rats (Fendt et al., 2008). Similarly, when a multiple-hit approach is applied, more attention should be paid to the timing of environmental insult and first and second order interactions among these hits, either gene or environment.
Animal models of schizophrenia utilizing multiple hits should have face (behavioral similarities, symptoms homology), construct (replicates pathology), and predictive (show pharmacological reversal of deficits or lack of pharmacological response) validity. Many of the models reviewed here have met these criteria and have shown that combining genetic and environmental risk factors or multiple developmental/environmental risk factors improves the model. Whether or not these models will offer better predictive validity for drug development is not yet known. The appeal of genetic and/or developmental models is the opportunity to intervene early in the progression of pathology and test potential preventive treatments. In several “single hit” models, particularly prenatal exposure to PolyI:C and neonatal ventral hippocampal lesion, preventative treatments have shown the ability to block some of the behavioral abnormalities, including reduced PPI (reviewed in Millan et al., 2016). In mice exposed to gestational PolyI:C, typical and atypical antipsychotics as well as antidepressants prevent PPI deficits in the model (Meyer et al., 2010). In the neonatal ventral hippocampal lesion model, the emergence of PPI deficits was prevented by adolescent treatment with antioxidants (Cabungcal et al., 2014). Perinatal NMDA antagonist (phencyclidine administration, PND 7–11)- induced PPI deficits were prevented by the mGlu5 positive allosteric modulator AD47273 and the nicotinic alpha-7 partial agonist, SSR180711 (Kjaerby et al., 2013). Preventive treatments have also been tested in a few two-hit models as well. In an effort to target the neuropathology in the model, pubertal treatment with minocycline prevented PPI deficits in mice exposed to MIA+juvenile stress (Giovanoli et al. 2016). In another study in inhibitory neuron-specific NR1 KO mice exposed to social isolation, chronic treatment with apocynin reversed PPI deficits in the mice (Jiang et al., 2013). Of course, none of these novel therapies have been shown to fully reverse the neuroanatomical and neurochemical deficits in individuals suffering from schizophrenia, but there is increasing evidence that neuroinflammation and oxidative stress may affect a subset of patients with schizophrenia and thus demonstrating efficacy in double hit models adds to our understanding of these putative risk factors and potential treatment approaches.
Another important point to consider in assessing models of risk factors for neuropsychiatric disease is that most of these risk factors, including both genetic and environmental, increase risk only moderately. For example, the estimated odds ratio for the exposure to obstetric complications increasing the risk of schizophrenia is 2.0, indicating a rather low relative risk associated with obstetric complications (Rapoport et al., 2005). Thus, it is not surprising that manipulating these risk factors on their own, or even in combination with one additional risk factor, does not produce profound behavioral alterations in model organisms. We must also realize that most environmental risk factors for schizophrenia are complex and occur in the context of other risk factors and/or protective factors, making them difficult to translate to animal models. For example, adolescents that smoke cannabis and are also socially isolated from peers may be more at risk than adolescents who smoke cannabis and have a supportive social network. Maternal infection combined with inadequate prenatal care may put the offspring more at risk than maternal infection combined with good prenatal care. The goal of animal models should not be to recapitulate the messy complexity and variability of the human condition, however. After all, the goal of animal research is to create simplified models to systematically manipulate variables of interest and control for as many other extraneous variables as possible. The hope is that these models will move beyond mere characterizations of behavioral/cognitive constructs deficient across neuropsychiatric disorders toward models with predictive power for drug development (Powell et al. 2012). Perhaps the combination of susceptibility genes and developmental risk factors will provide better models for medication development for neuropsychiatry. As our knowledge of the clinical condition improves, our models should attempt to more closely represent etiological risk factors and/or neuropathology in order to develop more predictive models for drug development (Moore, 2010). Combining risk factors, as reviewed here, moves the field toward developing these more refined models with potential for better translatability. We are well aware that all aspects of a heterogeneous disease will not be recreated in model organisms with a genetic mutation and/or developmental risk factors. Additionally, no single phenotype such as PPI is either necessary or sufficient to substantiate a model as having relevance to neuropsychiatric disease. Thus, as human studies materialize with more neurobiologically defined functional domains (as outlined in initiatives such as the NIH RDoC), translating these measures or “endophenotypes” to animal models will improve. As a preclinical behavioral measure, PPI has shown predictive validity in rodent pharmacological models, cross species homology, and sensitivity to genetic and neurodevelopmental risk factors for schizophrenia. Whether or not PPI has fulfilled its promise of informing clinical neuropsychiatry has been recently considered (Swerdlow et al., 2016; Swerdlow and Light, 2016). In terms of the double hit models reviewed here, PPI does not appear to be a sensitive measure of combined risk factors in many cases. Some double hit paradigms (e.g. MIA + adolescent stress), however, have shown synergistic effects on PPI that can be prevented by novel therapeutics, indicating a potentially enhanced ability to discover novel therapeutics (Giovanoli et al., 2016). While these rodent models are not without shortcomings, they are more closely approximating etiology by examining multiple risk factors. Hence, further consideration of environmental risk factors and systematic approaches to studying these combined risk factors would greatly benefit the genetic models. As the primary “hits” from GWAS studies emerge, future studies should focus on a sub-set of genes and either combine these candidate genes to examine epistatic interactions or, as reviewed here, combine candidate genes with environmental/developmental risk factors.
Table 2.
First hit | Model description/background/rationale | Second Hit | Effects on PPI | Prevention/Treatment strategies | References |
---|---|---|---|---|---|
Prenatal/Maternal Immune activation | |||||
Prenatal PolyI:C (1 mg/kg) GD 9 | In utero or maternal exposure with synthetic double stranded RNA PolyI:C or bacterial endotoxin LPS | Varied unpredictable stress between PND30–40 in C57BL/6 | Ø PPI in poly I:C vs Veh or Stressed vs NS. ↓PPI in polyI:C/Stressed vs polyI:C/NS. Significant ExE interaction | MIN (30mg/kg) before and during duration of unpredictable stress attenuated ↓PPI in ExE group. | Giovanoli et al, 2016 |
Prenatal PolyI:C (20 mg/kg) GD 12 | Juvenile restraint stress from PND33–35 in C57BL/6 | Ø PPI in poly I:C or Stressed vs controls. ↓PPI Poly I:C/stressed. Significant ExE interaction |
α-LA before each stress episode reversed ↓PPI in poly I:C/stressed mice | Deslauriers et al, 2013; Deslauriers et al 2014 | |
Prenatal PolyI:C (4 mg/kg) GD 15 | Juvenile varied stress from PND27–29 in Sprague Dawley rats | ↓PPI in poly I:C vs Sal. Ø PPI in stressed rats vs NS. Ø PPI in poly I:C/stressed vs Sal/NS. No ExE interaction | Yee et al, 2011 | ||
LPS (5μg/kg) on GD15/16 | Iron deficiency from GD2–PND7 | Ø PPI in LPS vs Sal. ↓PPI in ID/Sal vs IS/Sal and ID/LPS vs IS/Sal. No ExE interaction | Harvey et al, 2014 | ||
Post-weaning social isolation | |||||
Social isolation from PND21 in Wistar rats | Rodents raised singly housed in absence of any social interaction with other rats or mice immediately from weaning. | Escalating METH (0.2–6 mg/kg) on PND35–50 | ↓PPI in Iso vs Soc, ↓PPI in Iso/METH, Soc/METH Vs Soc/Veh. No ExE interaction |
Strauss et al, 2014 | |
Social isolation in Wistar rats on PND25 | Peripubertal administration of poly I:C (20 mg/kg) for 5 days from PND38–48 | ↓PPI in Iso/Sal vs Soc/Sal, Iso/poly I:C vs Soc/Sal on PND60. ↓PPI Iso/Sal vs Soc/Sal, Soc/poly I:C vs Soc/Sal & Iso/poly I:C vs Soc/Sal on PND80. Significant ExE interaction. | Lukasz et al, 2013 | ||
Neonatal NMDA antagonist/Neurotoxin | |||||
Neonatal MK-801 (0.2 mg/kg) from PND7–10 in Sprague Dawley rats | Neonatal NMDA antagonist exposure to model hypothesized NMDA hypofunction in schizophrenia and as a general neonatal insult | Social isolation from PND21 | ↓PPI in Iso vs Soc on PND77&91. ↓PPI in MK-801/Iso vs Soc/Veh on all intensities at PND91. | CLO injection before PPI on PND84 attenuate ↓PPI in MK801/isolates vs social/veh | Lim et al, 2012 |
Neonatal PCP (10mg/kg) on PND7,9,11 in Lister-hooded rats | Social isolation from PND23 | Ø PPI in Iso vs Soc, PCP/Soc vs PCP/Veh. ↓PPI in PCP/Iso vs Veh/Soc. Significant ExE interaction | Gaskin et al, 2014 | ||
Neonatal DOM injections from PND8-14 in Sprague-Dawley rats | Domoic acid (DOM) is a kainite agonist which produces neurotoxicity | Social isolation from PND21 for 12 weeks | ↓PPI in Iso vs Soc. ↑PPI in Iso/DOM vs Iso/Sal. No ExE interaction | Marriott et al, 2016 | |
Maternal separation | |||||
Maternal separation from PND 3–10 in Sprague-Dawley rats | Rodents deprived perinatally from stimulations from dam and littermates. | Avoidance conditioning (PND49–56) and immediately injected with PCP after every conditioning session | Ø PPI in MS vs Non-MS. Effects of PCP and conditioned avoidance are not clearly documented | Chen et al 2011 | |
Maternal separation on PND 9 for 24h in Wistar rats | Adolescent Corticosterone treatment at 8 weeks of age | Ø PPI in MS vs Non-MS. AMPH & APO ↓ PPI in Non-MS & Non-MS/CORT. APO ↓ PPI in MS. Ø PPI in MS by AMPH. Ø PPI by APO & AMPH in MS/CORT. DPAT ↓ PPI in Non-MS, Non-MS CORT, MS & MS/CORT | Choy et al 2008 |
Abbreviations: AMPH amphetamine, APO apomorphine, CLO clozapine, DOM domoic acid, DPAT 8-hydroxy-dipropylaminotetralin, E environment, EE environmental enrichment, F female, GD gestational day, ISI interstimulus interval, Iso isolate, IS iron sufficient, ID Iron deficient, LA lipoic acid, LPS lipopolysaccharide, M male, NS non-stressed, MIN minocycline, MS maternal separation, METH methamphetamine, NMDA N-Methyl-D-aspartate, PA magnitude of response to pulse alone, PCP phencyclidine, PND postnatal day, PNS prenatal stress, Poly I:C polyinosinic: polycytidylic acid, PP prepulse, PPI prepulse inhibition of startle, SI social isolation, Soc socials, SZ schizophrenia, Veh vehicle, ↓ decreased, ↑ increased, Ø unchanged
Acknowledgments
We thank Drs. Victoria Risbrough, Xianjin Zhou, and Tiffany Greenwood for helpful discussions and comments. This work was supported by ES025585 and by the Veterans Affairs VISN 22 Mental Illness Research, Education, and Clinical Center.
This work was supported by ES025585 and Veteran’s Affairs VISN 22 MIRECC.
Funding Source Declaration: NIH grant ES025585 and Veteran’s Affairs VISN 22 MIRECC supported the authors during the writing of this manuscript.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of Interest Statement: There are no financial/personal interests or beliefs that could affect the objectivity of the authors.
Contributors: Asma Khan and Susan Powell both wrote the manuscript.
Author Agreement/Declaration: I certify that all authors (Asma Khan and myself, Susan Powell) have seen and approved the final version of the manuscript being submitted. I warrant that this article is our (the authors’) original work, hasn’t received prior publication and isn’t under consideration for publication elsewhere.
References
- Aas M, Djurovic S, Athanasiu L, Steen NE, Agartz I, Lorentzen S, Sundet K, Andreassen OA, Melle I. Serotonin transporter gene polymorphism, childhood trauma, and cognition in patients with psychotic disorders. Schizophr Bull. 2012;38(1):15–22. doi: 10.1093/schbul/sbr113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Abazyan B, Dziedzic J, Hua K, Abazyan S, Yang C, Mori S, Pletnikov MV, Guilarte TR. Chronic Exposure of Mutant DISC1 Mice to Lead Produces Sex-Dependent Abnormalities Consistent With Schizophrenia and Related Mental Disorders: A Gene-Environment Interaction Study. Schizophrenia Bulletin. 2014;40(3):575–584. doi: 10.1093/schbul/sbt071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Addington J, Penn D, Woods SW, Addington D, Perkins DO. Social functioning in individuals at clinical high risk for psychosis. Schizophr Res. 2008;99(1–3):119–124. doi: 10.1016/j.schres.2007.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ahmari SE, Risbrough VB, Geyer MA, Simpson HB. Impaired sensorimotor gating in unmedicated adults with obsessive-compulsive disorder. Neuropsychopharmacology. 2012;37(5):1216–1223. doi: 10.1038/npp.2011.308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ahmari SE, Risbrough VB, Geyer MA, Simpson HB. Prepulse Inhibition Deficits in Obsessive-Compulsive Disorder are More Pronounced in Females. Neuropsychopharmacology. 2016;41(13):2963–2964. doi: 10.1038/npp.2015.363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alemany S, Arias B, Aguilera M, Villa H, Moya J, Ibanez MI, Vossen H, Gasto C, Ortet G, Fananas L. Childhood abuse, the BDNF-Val66Met polymorphism and adult psychotic-like experiences. Br J Psychiatry. 2011;199(1):38–42. doi: 10.1192/bjp.bp.110.083808. [DOI] [PubMed] [Google Scholar]
- Alemany S, Arias B, Fatjo-Vilas M, Villa H, Moya J, Ibanez MI, Ortet G, Gasto C, Fananas L. Psychosis-inducing effects of cannabis are related to both childhood abuse and COMT genotypes. Acta Psychiatr Scand. 2014;129(1):54–62. doi: 10.1111/acps.12108. [DOI] [PubMed] [Google Scholar]
- Andréasson S, Engström A, Allebeck P, Rydberg U. CANNABIS AND SCHIZOPHRENIA A Longitudinal Study of Swedish Conscripts. The Lancet. 1987;330(8574):1483–1486. doi: 10.1016/s0140-6736(87)92620-1. [DOI] [PubMed] [Google Scholar]
- Arguello PA, Gogos JA. Cognition in mouse models of schizophrenia susceptibility genes. Schizophr Bull. 2010;36(2):289–300. doi: 10.1093/schbul/sbp153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arguello PA, Gogos JA. Psychiatric genetics and the generation of mutant animal models. In: O’Donnell P, editor. Animal Models of Schizophrenia and Related Disorders. Springer; New York: 2011. pp. 195–215. [Google Scholar]
- Arseneault L, Cannon M, Poulton R, Murray R, Caspi A, Moffitt TE. Cannabis use in adolescence and risk for adult psychosis: longitudinal prospective study. BMJ. 2002;325(7374):1212–1213. doi: 10.1136/bmj.325.7374.1212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. The international journal of neuropsychopharmacology. 2010;13(4):475–485. doi: 10.1017/S1461145709990253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Autry AE, Monteggia LM. Brain-Derived Neurotrophic Factor and Neuropsychiatric Disorders. Pharmacological Reviews. 2012;64(2):238–258. doi: 10.1124/pr.111.005108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ayhan Y, McFarland R, Pletnikov MV. Animal models of gene-environment interaction in schizophrenia: A dimensional perspective. Prog Neurobiol. 2016;136:1–27. doi: 10.1016/j.pneurobio.2015.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barnes TR, Mutsatsa SH, Hutton SB, Watt HC, Joyce EM. Comorbid substance use and age at onset of schizophrenia. Br J Psychiatry. 2006;188:237–242. doi: 10.1192/bjp.bp.104.007237. [DOI] [PubMed] [Google Scholar]
- Berger UV, Luthi-Carter R, Passani LA, Elkabes S, Black I, Konradi C, Coyle JT. Glutamate carboxypeptidase II is expressed by astrocytes in the adult rat nervous system. J Comp Neurol. 1999;415(1):52–64. doi: 10.1002/(sici)1096-9861(19991206)415:1<52::aid-cne4>3.0.co;2-k. [DOI] [PubMed] [Google Scholar]
- Blackwood DH, Fordyce A, Walker MT, St Clair DM, Porteous DJ, Muir WJ. Schizophrenia and affective disorders–cosegregation with a translocation at chromosome 1q42 that directly disrupts brain-expressed genes: clinical and P300 findings in a family. Am J Hum Genet. 2001;69(2):428–433. doi: 10.1086/321969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boksa P. Animal models of obstetric complications in relation to schizophrenia. Brain Res Brain Res Rev. 2004;45(1):1–17. doi: 10.1016/j.brainresrev.2004.01.001. [DOI] [PubMed] [Google Scholar]
- Borglum AD, Demontis D, Grove J, Pallesen J, Hollegaard MV, Pedersen CB, Hedemand A, Mattheisen M, Uitterlinden A, Nyegaard M, Orntoft T, Wiuf C, Didriksen M, Nordentoft M, Nothen MM, Rietschel M, Ophoff RA, Cichon S, Yolken RH, Hougaard DM, Mortensen PB, Mors O. Genome-wide study of association and interaction with maternal cytomegalovirus infection suggests new schizophrenia loci. Mol Psychiatry. 2014;19(3):325–333. doi: 10.1038/mp.2013.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bortolato M, Aru GN, Frau R, Orru M, Luckey GC, Boi G, Gessa GL. The CB receptor agonist WIN 55,212-2 fails to elicit disruption of prepulse inhibition of the startle in Sprague-Dawley rats. Psychopharmacology (Berl) 2005;177(3):264–271. doi: 10.1007/s00213-004-1941-4. [DOI] [PubMed] [Google Scholar]
- Bortolato M, Bini V, Frau R, Devoto P, Pardu A, Fan Y, Solbrig MV. Juvenile cannabinoid treatment induces frontostriatal gliogenesis in Lewis rats. European Neuropsychopharmacology. 2014;24(6):974–985. doi: 10.1016/j.euroneuro.2013.12.011. [DOI] [PubMed] [Google Scholar]
- Boucher AA, Arnold JC, Duffy L, Schofield PR, Micheau J, Karl T. Heterozygous neuregulin 1 mice are more sensitive to the behavioural effects of Δ9-tetrahydrocannabinol. Psychopharmacology. 2007;192(3):325–336. doi: 10.1007/s00213-007-0721-3. [DOI] [PubMed] [Google Scholar]
- Boucher AA, Hunt GE, Micheau J, Huang X, McGregor IS, Karl T, Arnold JC. The schizophrenia susceptibility gene neuregulin 1 modulates tolerance to the effects of cannabinoids. International Journal of Neuropsychopharmacology. 2011;14(5):631–643. doi: 10.1017/S146114571000091X. [DOI] [PubMed] [Google Scholar]
- Braff DL. Prepulse inhibition of the startle reflex: a window on the brain in schizophrenia. Curr Top Behav Neurosci. 2010;4:349–371. doi: 10.1007/7854_2010_61. [DOI] [PubMed] [Google Scholar]
- Braff DL. Gating in schizophrenia: from genes to cognition (to real world function?) Biol Psychiatry. 2011;69(5):395–396. doi: 10.1016/j.biopsych.2011.01.002. [DOI] [PubMed] [Google Scholar]
- Braff DL, Geyer MA, Swerdlow NR. Human studies of prepulse inhibition of startle: normal subjects, patient groups, and pharmacological studies. Psychopharmacology (Berl) 2001;156(2–3):234–258. doi: 10.1007/s002130100810. [DOI] [PubMed] [Google Scholar]
- Braff DL, Grillon C, Geyer MA. Gating and habituation of the startle reflex in schizophrenic patients. Arch Gen Psychiatry. 1992;49(3):206–215. doi: 10.1001/archpsyc.1992.01820030038005. [DOI] [PubMed] [Google Scholar]
- Braff DL, Light GA, Swerdlow NR. Prepulse inhibition and P50 suppression are both deficient but not correlated in schizophrenia patients. Biol Psychiatry. 2007;61(10):1204–1207. doi: 10.1016/j.biopsych.2006.08.015. [DOI] [PubMed] [Google Scholar]
- Brandon NJ, Sawa A. Linking neurodevelopmental and synaptic theories of mental illness through DISC1. Nature reviews Neuroscience. 2011;12(12):707–722. doi: 10.1038/nrn3120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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(8):774–780. doi: 10.1001/archpsyc.61.8.774. [DOI] [PubMed] [Google Scholar]
- 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(5):889–895. doi: 10.1176/appi.ajp.161.5.889. [DOI] [PubMed] [Google Scholar]
- 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(4):767–773. doi: 10.1176/appi.ajp.162.4.767. [DOI] [PubMed] [Google Scholar]
- Brown AS, Susser ES. In utero infection and adult schizophrenia. Ment Retard Dev Disabil Res Rev. 2002;8(1):51–57. doi: 10.1002/mrdd.10004. [DOI] [PubMed] [Google Scholar]
- Brown AS, Susser ES. Prenatal nutritional deficiency and risk of adult schizophrenia. Schizophr Bull. 2008;34(6):1054–1063. doi: 10.1093/schbul/sbn096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Buka SL, Cannon TD, Torrey EF, Yolken RH. Maternal exposure to herpes simplex virus and risk of psychosis among adult offspring. Biol Psychiatry. 2008;63(8):809–815. doi: 10.1016/j.biopsych.2007.09.022. [DOI] [PubMed] [Google Scholar]
- 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(4):411–420. doi: 10.1006/brbi.2001.0644. [DOI] [PubMed] [Google Scholar]
- 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(2):549–555. doi: 10.1016/j.bbr.2004.03.023. [DOI] [PubMed] [Google Scholar]
- 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(4):651–655. doi: 10.1016/j.physbeh.2004.03.004. [DOI] [PubMed] [Google Scholar]
- 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(1):119–124. doi: 10.1016/j.bbr.2006.07.015. [DOI] [PubMed] [Google Scholar]
- Burt MA, Ryan CL, Doucette TA. Altered responses to novelty and drug reinforcement in adult rats treated neonatally with domoic acid. Physiology & behavior. 2008a;93(1–2):327–336. doi: 10.1016/j.physbeh.2007.09.003. [DOI] [PubMed] [Google Scholar]
- Burt MA, Ryan CL, Doucette TA. Low dose domoic acid in neonatal rats abolishes nicotine induced conditioned place preference during late adolescence. Amino acids. 2008b;35(1):247–249. doi: 10.1007/s00726-007-0584-2. [DOI] [PubMed] [Google Scholar]
- Buse J, Beste C, Herrmann E, Roessner V. Neural correlates of altered sensorimotor gating in boys with Tourette Syndrome: A combined EMG/fMRI study. The World Journal of Biological Psychiatry. 2016;17(3):187–197. doi: 10.3109/15622975.2015.1112033. [DOI] [PubMed] [Google Scholar]
- Cabungcal JH, Counotte DS, Lewis EM, Tejeda HA, Piantadosi P, Pollock C, Calhoon GG, Sullivan EM, Presgraves E, Kil J, Hong LE, Cuenod M, Do KQ, O’Donnell P. Juvenile antioxidant treatment prevents adult deficits in a developmental model of schizophrenia. Neuron. 2014;83(5):1073–1084. doi: 10.1016/j.neuron.2014.07.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cadenhead KS. Startle reactivity and prepulse inhibition in prodromal and early psychosis: Effects of age, antipsychotics, tobacco and cannabis in a vulnerable population. Psychiatry Research. 2011;188(2):208–216. doi: 10.1016/j.psychres.2011.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cadenhead KS, Geyer MA, Braff DL. Impaired startle prepulse inhibition and habituation in patients with schizotypal personality disorder. Am J Psychiatry. 1993;150(12):1862–1867. doi: 10.1176/ajp.150.12.1862. [DOI] [PubMed] [Google Scholar]
- Cadenhead KS, Swerdlow NR, Shafer KM, Diaz M, Braff DL. Modulation of the startle response and startle laterality in relatives of schizophrenic patients and in subjects with schizotypal personality disorder: evidence of inhibitory deficits. Am J Psychiatry. 2000;157(10):1660–1668. doi: 10.1176/appi.ajp.157.10.1660. [DOI] [PubMed] [Google Scholar]
- Cannon M, Jones PB, Murray RM. Obstetric complications and schizophrenia: historical and meta-analytic review. Am J Psychiatry. 2002;159(7):1080–1092. doi: 10.1176/appi.ajp.159.7.1080. [DOI] [PubMed] [Google Scholar]
- 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(1):28–37. doi: 10.1001/archgenpsychiatry.2007.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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(4):653–669. doi: 10.1093/oxfordjournals.schbul.a007037. [DOI] [PubMed] [Google Scholar]
- Cannon TD, Yu C, Addington J, Bearden CE, Cadenhead KS, Cornblatt BA, Heinssen R, Jeffries CD, Mathalon DH, McGlashan TH, Perkins DO, Seidman LJ, Tsuang MT, Walker EF, Woods SW, Kattan MW. An Individualized Risk Calculator for Research in Prodromal Psychosis. Am J Psychiatry. 2016;173(10):980–988. doi: 10.1176/appi.ajp.2016.15070890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Casey BJ, Jones RM, Hare TA. The adolescent brain. Ann N Y Acad Sci. 2008;1124:111–126. doi: 10.1196/annals.1440.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Caspi A, Moffitt TE, Cannon M, McClay J, Murray R, Harrington H, Taylor A, Arseneault L, Williams B, Braithwaite A, Poulton R, Craig IW. Moderation of the effect of adolescent-onset cannabis use on adult psychosis by a functional polymorphism in the catechol-O-methyltransferase gene: longitudinal evidence of a gene × environment interaction. Biol Psychiatry. 2005;57(10):1117–1127. doi: 10.1016/j.biopsych.2005.01.026. [DOI] [PubMed] [Google Scholar]
- Cassidy AW, Mulvany SK, Pangalos MN, Murphy KJ, Regan CM. Developmental emergence of reelin deficits in the prefrontal cortex of Wistar rats reared in social isolation. Neuroscience. 2010a;166(2):377–385. doi: 10.1016/j.neuroscience.2009.12.045. [DOI] [PubMed] [Google Scholar]
- Cassidy AW, Mulvany SK, Pangalos MN, Murphy KJ, Regan CM. Reduced reelin protein synthesis in ventral hippocampus of isolation reared Wistar rats accompanies impaired avoidance conditioning. Behavioural Brain Research. 2010b;213(1):130–134. doi: 10.1016/j.bbr.2010.04.040. [DOI] [PubMed] [Google Scholar]
- Castellanos FX, Fine EJ, Kaysen D, Marsh WL, Rapoport JL, Hallett M. Sensorimotor gating in boys with Tourette’s syndrome and ADHD: preliminary results. Biol Psychiatry. 1996;39(1):33–41. doi: 10.1016/0006-3223(95)00101-8. [DOI] [PubMed] [Google Scholar]
- Chen J, Wang Z, Li M. Multiple ‘hits’ during postnatal and early adulthood periods disrupt the normal development of sensorimotor gating ability in rats. Journal of psychopharmacology (Oxford, England) 2011;25(3):379–392. doi: 10.1177/0269881109354929. [DOI] [PubMed] [Google Scholar]
- Chen YA, Scheller RH. SNARE-mediated membrane fusion. Nat Rev Mol Cell Biol. 2001;2(2):98–106. doi: 10.1038/35052017. [DOI] [PubMed] [Google Scholar]
- Cheslack-Postava K, Brown AS, Chudal R, Suominen A, Huttunen J, Surcel HM, Sourander A. Maternal exposure to sexually transmitted infections and schizophrenia among offspring. Schizophr Res. 2015;166(1–3):255–260. doi: 10.1016/j.schres.2015.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Choy KH, de Visser YP, van den Buuse M. The effect of ‘two hit’ neonatal and young-adult stress on dopaminergic modulation of prepulse inhibition and dopamine receptor density. Br J Pharmacol. 2009;156(2):388–396. doi: 10.1111/j.1476-5381.2008.00008.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Choy KH, van den Buuse M. Attenuated disruption of prepulse inhibition by dopaminergic stimulation after maternal deprivation and adolescent corticosterone treatment in rats. Eur Neuropsychopharmacol. 2008;18(1):1–13. doi: 10.1016/j.euroneuro.2007.03.011. [DOI] [PubMed] [Google Scholar]
- Coleman LG, Jr, He J, Lee J, Styner M, Crews FT. Adolescent Binge Drinking Alters Adult Brain Neurotransmitter Gene Expression, Behavior, Brain Regional Volumes, and Neurochemistry in Mice. Alcoholism: Clinical and Experimental Research. 2011;35(4):671–688. doi: 10.1111/j.1530-0277.2010.01385.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Collip D, Myin-Germeys I, Wichers M, Jacobs N, Derom C, Thiery E, Lataster T, Simons C, Delespaul P, Marcelis M, van Os J, van Winkel R. FKBP5 as a possible moderator of the psychosis-inducing effects of childhood trauma. Br J Psychiatry. 2013;202(4):261–268. doi: 10.1192/bjp.bp.112.115972. [DOI] [PubMed] [Google Scholar]
- Consortium, S.W.G.o.t.P.G. Biological insights from 108 schizophrenia-associated genetic loci. Nature. 2014;511(7510):421–427. doi: 10.1038/nature13595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cuthbert BN, Insel TR. Toward the future of psychiatric diagnosis: the seven pillars of RDoC. BMC Med. 2013;11:126. doi: 10.1186/1741-7015-11-126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davidsson P, Gottfries J, Bogdanovic N, Ekman R, Karlsson I, Gottfries CG, Blennow K. The synaptic-vesicle-specific proteins rab3a and synaptophysin are reduced in thalamus and related cortical brain regions in schizophrenic brains. Schizophr Res. 1999;40(1):23–29. doi: 10.1016/s0920-9964(99)00037-7. [DOI] [PubMed] [Google Scholar]
- Davis J, Eyre H, Jacka FN, Dodd S, Dean O, McEwen S, Debnath M, McGrath J, Maes M, Amminger P, McGorry PD, Pantelis C, Berk M. A review of vulnerability and risks for schizophrenia: Beyond the two hit hypothesis. Neuroscience & Biobehavioral Reviews. 2016;65:185–194. doi: 10.1016/j.neubiorev.2016.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Sousa KR, Tiwari AK, Giuffra DE, Mackenzie B, Zai CC, Kennedy JL. Age at onset of schizophrenia: cannabis, COMT gene, and their interactions. Schizophr Res. 2013;151(1–3):289–290. doi: 10.1016/j.schres.2013.10.037. [DOI] [PubMed] [Google Scholar]
- Dean K, Bramon E, Murray RM. The causes of schizophrenia: neurodevelopment and other risk factors. J Psychiatr Pract. 2003;9(6):442–454. doi: 10.1097/00131746-200311000-00007. [DOI] [PubMed] [Google Scholar]
- Demontis D, Nyegaard M, Buttenschon HN, Hedemand A, Pedersen CB, Grove J, Flint TJ, Nordentoft M, Werge T, Hougaard DM, Sorensen KM, Yolken RH, Mors O, Borglum AD, Mortensen PB. Association of GRIN1 and GRIN2A-D with schizophrenia and genetic interaction with maternal herpes simplex virus-2 infection affecting disease risk. Am J Med Genet B Neuropsychiatr Genet. 2011;156B(8):913–922. doi: 10.1002/ajmg.b.31234. [DOI] [PubMed] [Google Scholar]
- Deslauriers J, Larouche A, Sarret P, Grignon S. Combination of prenatal immune challenge and restraint stress affects prepulse inhibition and dopaminergic/GABAergic markers. Progress in neuro-psychopharmacology & biological psychiatry. 2013;45:156–164. doi: 10.1016/j.pnpbp.2013.05.006. [DOI] [PubMed] [Google Scholar]
- Deslauriers J, Racine W, Sarret P, Grignon S. Preventive effect of alpha-lipoic acid on prepulse inhibition deficits in a juvenile two-hit model of schizophrenia. Neuroscience. 2014;272:261–270. doi: 10.1016/j.neuroscience.2014.04.061. [DOI] [PubMed] [Google Scholar]
- Devlin AM, Ling EH, Peerson JM, Fernando S, Clarke R, Smith AD, Halsted CH. Glutamate carboxypeptidase II: a polymorphism associated with lower levels of serum folate and hyperhomocysteinemia. Hum Mol Genet. 2000;9(19):2837–2844. doi: 10.1093/hmg/9.19.2837. [DOI] [PubMed] [Google Scholar]
- Di Forti M, Iyegbe C, Sallis H, Kolliakou A, Falcone MA, Paparelli A, Sirianni M, La Cascia C, Stilo SA, Marques TR, Handley R, Mondelli V, Dazzan P, Pariante C, David AS, Morgan C, Powell J, Murray RM. Confirmation that the AKT1 (rs2494732) genotype influences the risk of psychosis in cannabis users. Biol Psychiatry. 2012;72(10):811–816. doi: 10.1016/j.biopsych.2012.06.020. [DOI] [PubMed] [Google Scholar]
- Donoghue K, Doody GA, Murray RM, Jones PB, Morgan C, Dazzan P, Hart J, Mazzoncini R, Maccabe JH. Cannabis use, gender and age of onset of schizophrenia: data from the AESOP study. Psychiatry Res. 2014;215(3):528–532. doi: 10.1016/j.psychres.2013.12.038. [DOI] [PubMed] [Google Scholar]
- Eells JB, Lipska BK, Yeung SK, Misler JA, Nikodem VM. Nurr1-null heterozygous mice have reduced mesolimbic and mesocortical dopamine levels and increased stress-induced locomotor activity. Behav Brain Res. 2002;136(1):267–275. doi: 10.1016/s0166-4328(02)00185-7. [DOI] [PubMed] [Google Scholar]
- Eells JB, Misler JA, Nikodem VM. Early postnatal isolation reduces dopamine levels, elevates dopamine turnover and specifically disrupts prepulse inhibition in Nurr1-null heterozygous mice. Neuroscience. 2006;140(4):1117–1126. doi: 10.1016/j.neuroscience.2005.12.065. [DOI] [PubMed] [Google Scholar]
- Eells JB, Varela-Stokes A, Guo-Ross SX, Kummari E, Smith HM, Cox E, Lindsay DS. Chronic Toxoplasma gondii in Nurr1-Null Heterozygous Mice Exacerbates Elevated Open Field Activity. PLOS ONE. 2015;10(4):e0119280. doi: 10.1371/journal.pone.0119280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ellenbroek BA, Cools AR. The long-term effects of maternal deprivation depend on the genetic background. Neuropsychopharmacology. 2000;23(1):99–106. doi: 10.1016/S0893-133X(00)00088-9. [DOI] [PubMed] [Google Scholar]
- 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(3):251–260. doi: 10.1016/s0920-9964(97)00149-7. [DOI] [PubMed] [Google Scholar]
- Estes ML, McAllister AK. Maternal immune activation: Implications for neuropsychiatric disorders. Science. 2016;353(6301):772–777. doi: 10.1126/science.aag3194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eyles DW, Burne TH, McGrath JJ. Vitamin D, effects on brain development, adult brain function and the links between low levels of vitamin D and neuropsychiatric disease. Front Neuroendocrinol. 2013;34(1):47–64. doi: 10.1016/j.yfrne.2012.07.001. [DOI] [PubMed] [Google Scholar]
- 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 doi: 10.1016/j.psyneuen.2009.04.015. [DOI] [PubMed] [Google Scholar]
- Fatemi SH, Folsom TD. The Neurodevelopmental Hypothesis of Schizophrenia, Revisited. Schizophr Bull. 2009;35(3):528–548. doi: 10.1093/schbul/sbn187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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(4):138–145. doi: 10.1055/s-2008-1058107. [DOI] [PubMed] [Google Scholar]
- Finamore TL, Port RL. Developmental stress disrupts habituation but spares prepulse inhibition in young rats. Physiol Behav. 2000;69(4–5):527–530. doi: 10.1016/s0031-9384(00)00205-5. [DOI] [PubMed] [Google Scholar]
- Frankland PW, Wang Y, Rosner B, Shimizu T, Balleine BW, Dykens EM, Ornitz EM, Silva AJ. Sensorimotor gating abnormalities in young males with fragile × syndrome and Fmr1-knockout mice. Mol Psychiatry. 2004;9(4):417–425. doi: 10.1038/sj.mp.4001432. [DOI] [PubMed] [Google Scholar]
- Fridlund AJ, Cacioppo JT. Guidelines for human electromyographic research. Psychophysiology. 1986;23(5):567–589. doi: 10.1111/j.1469-8986.1986.tb00676.x. [DOI] [PubMed] [Google Scholar]
- Frost WN, Tian LM, Hoppe TA, Mongeluzi DL, Wang J. A cellular mechanism for prepulse inhibition. Neuron. 2003;40(5):991–1001. doi: 10.1016/s0896-6273(03)00731-1. [DOI] [PubMed] [Google Scholar]
- Gage SH, Hickman M, Zammit S. Association Between Cannabis and Psychosis: Epidemiologic Evidence. Biological Psychiatry. 2016;79(7):549–556. doi: 10.1016/j.biopsych.2015.08.001. [DOI] [PubMed] [Google Scholar]
- Gaskin PLR, Alexander SPH, Fone KCF. Neonatal phencyclidine administration and post-weaning social isolation as a dual-hit model of ‘schizophrenia-like’ behaviour in the rat. Psychopharmacology. 2014;231(12):2533–2545. doi: 10.1007/s00213-013-3424-y. [DOI] [PubMed] [Google Scholar]
- Geyer MA, McIlwain KL, Paylor R. Mouse genetic models for prepulse inhibition: an early review. Mol Psychiatry. 2002;7(10):1039–1053. doi: 10.1038/sj.mp.4001159. [DOI] [PubMed] [Google Scholar]
- Giakoumaki SG, Roussos P, Bitsios P. Improvement of prepulse inhibition and executive function by the COMT inhibitor tolcapone depends on COMT Val158Met polymorphism. Neuropsychopharmacology. 2008;33(13):3058–3068. doi: 10.1038/npp.2008.82. [DOI] [PubMed] [Google Scholar]
- Giedd JN. The teen brain: insights from neuroimaging. J Adolesc Health. 2008;42(4):335–343. doi: 10.1016/j.jadohealth.2008.01.007. [DOI] [PubMed] [Google Scholar]
- Giovanoli S, Engler H, Engler A, Richetto J, Feldon J, Riva MA, Schedlowski M, Meyer U. Preventive effects of minocycline in a neurodevelopmental two-hit model with relevance to schizophrenia. Transl Psychiatry. 2016;6:e772. doi: 10.1038/tp.2016.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Giovanoli S, Engler H, Engler A, Richetto J, Voget M, Willi R, Winter C, Riva MA, Mortensen PB, Schedlowski M, Meyer U. Stress in puberty unmasks latent neuropathological consequences of prenatal immune activation in mice. Science. 2013;339(6123):1095–1099. doi: 10.1126/science.1228261. [DOI] [PubMed] [Google Scholar]
- Girardet C, Lebrun B, Cabirol-Pol MJ, Tardivel C, Francois-Bellan AM, Becquet D, Bosler O. Brain-derived neurotrophic factor/TrkB signaling regulates daily astroglial plasticity in the suprachiasmatic nucleus: electron-microscopic evidence in mouse. Glia. 2013;61(7):1172–1177. doi: 10.1002/glia.22509. [DOI] [PubMed] [Google Scholar]
- Gottesman II. Schizophrenia Genesis. W.H. Freeman; New York: 1991. [Google Scholar]
- Graham FK. Presidential Address, 1974. The more or less startling effects of weak prestimulation. Psychophysiology. 1975;12(3):238–248. doi: 10.1111/j.1469-8986.1975.tb01284.x. [DOI] [PubMed] [Google Scholar]
- Greenwood TA, Braff DL, Light GA, Cadenhead KS, Calkins ME, Dobie DJ, Freedman R, Green MF, Gur RE, Gur RC, Mintz J, Nuechterlein KH, Olincy A, Radant AD, Seidman LJ, Siever LJ, Silverman JM, Stone WS, Swerdlow NR, Tsuang DW, Tsuang MT, Turetsky BI, Schork NJ. Initial heritability analyses of endophenotypic measures for schizophrenia: the consortium on the genetics of schizophrenia. Arch Gen Psychiatry. 2007;64(11):1242–1250. doi: 10.1001/archpsyc.64.11.1242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greenwood TA, Lazzeroni LC, Murray SS, Cadenhead KS, Calkins ME, Dobie DJ, Green MF, Gur RE, Gur RC, Hardiman G, Kelsoe JR, Leonard S, Light GA, Nuechterlein KH, Olincy A, Radant AD, Schork NJ, Seidman LJ, Siever LJ, Silverman JM, Stone WS, Swerdlow NR, Tsuang DW, Tsuang MT, Turetsky BI, Freedman R, Braff DL. Analysis of 94 candidate genes and 12 endophenotypes for schizophrenia from the Consortium on the Genetics of Schizophrenia. Am J Psychiatry. 2011;168(9):930–946. doi: 10.1176/appi.ajp.2011.10050723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greenwood TA, Light GA, Swerdlow NR, Radant AD, Braff DL. Association analysis of 94 candidate genes and schizophrenia-related endophenotypes. PLoS One. 2012;7(1):e29630. doi: 10.1371/journal.pone.0029630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greenwood TA, Swerdlow NR, Gur RE, Cadenhead KS, Calkins ME, Dobie DJ, Freedman R, Green MF, Gur RC, Lazzeroni LC, Nuechterlein KH, Olincy A, Radant AD, Ray A, Schork NJ, Seidman LJ, Siever LJ, Silverman JM, Stone WS, Sugar CA, Tsuang DW, Tsuang MT, Turetsky BI, Light GA, Braff DL. Genome-wide linkage analyses of 12 endophenotypes for schizophrenia from the Consortium on the Genetics of Schizophrenia. Am J Psychiatry. 2013;170(5):521–532. doi: 10.1176/appi.ajp.2012.12020186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guloksuz S, van Nierop M, Lieb R, van Winkel R, Wittchen HU, van Os J. Evidence that the presence of psychosis in non-psychotic disorder is environment-dependent and mediated by severity of non-psychotic psychopathology. Psychol Med. 2015;45(11):2389–2401. doi: 10.1017/S0033291715000380. [DOI] [PubMed] [Google Scholar]
- Haque FN, Lipina TV, Roder JC, Wong AHC. Social defeat interacts with Disc1 mutations in the mouse to affect behavior. Behavioural Brain Research. 2012;233(2):337–344. doi: 10.1016/j.bbr.2012.05.037. [DOI] [PubMed] [Google Scholar]
- Harley M, Kelleher I, Clarke M, Lynch F, Arseneault L, Connor D, Fitzpatrick C, Cannon M. Cannabis use and childhood trauma interact additively to increase the risk of psychotic symptoms in adolescence. Psychol Med. 2010;40(10):1627–1634. doi: 10.1017/S0033291709991966. [DOI] [PubMed] [Google Scholar]
- Harrison PJ, Law AJ. Neuregulin 1 and schizophrenia: genetics, gene expression, and neurobiology. Biol Psychiatry. 2006;60(2):132–140. doi: 10.1016/j.biopsych.2005.11.002. [DOI] [PubMed] [Google Scholar]
- Hashimoto H, Hashimoto R, Shintani N, Tanaka K, Yamamoto A, Hatanaka M, Guo X, Morita Y, Tanida M, Nagai K, Takeda M, Baba A. Depression-like behavior in the forced swimming test in PACAP-deficient mice: amelioration by the atypical antipsychotic risperidone. Journal of neurochemistry. 2009;110(2):595–602. doi: 10.1111/j.1471-4159.2009.06168.x. [DOI] [PubMed] [Google Scholar]
- Hashimoto H, Shintani N, Tanaka K, Mori W, Hirose M, Matsuda T, Sakaue M, Miyazaki J, Niwa H, Tashiro F, Yamamoto K, Koga K, Tomimoto S, Kunugi A, Suetake S, Baba A. Altered psychomotor behaviors in mice lacking pituitary adenylate cyclase-activating polypeptide (PACAP) Proceedings of the National Academy of Sciences of the United States of America. 2001;98(23):13355–13360. doi: 10.1073/pnas.231094498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hashimoto R, Hashimoto H, Shintani N, Chiba S, Hattori S, Okada T, Nakajima M, Tanaka K, Kawagishi N, Nemoto K, Mori T, Ohnishi T, Noguchi H, Hori H, Suzuki T, Iwata N, Ozaki N, Nakabayashi T, Saitoh O, Kosuga A, Tatsumi M, Kamijima K, Weinberger DR, Kunugi H, Baba A. Pituitary adenylate cyclase-activating polypeptide is associated with schizophrenia. Mol Psychiatry. 2007;12(11):1026–1032. doi: 10.1038/sj.mp.4001982. [DOI] [PubMed] [Google Scholar]
- Hessl D, Berry-Kravis E, Cordeiro L, Yuhas J, Ornitz EM, Campbell A, Chruscinski E, Hervey C, Long JM, Hagerman RJ. Prepulse inhibition in fragile × syndrome: feasibility, reliability, and implications for treatment. Am J Med Genet B Neuropsychiatr Genet. 2009;150B(4):545–553. doi: 10.1002/ajmg.b.30858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoenig K, Hochrein A, Quednow BB, Maier W, Wagner M. Impaired prepulse inhibition of acoustic startle in obsessive-compulsive disorder. Biol Psychiatry. 2005;57(10):1153–1158. doi: 10.1016/j.biopsych.2005.01.040. [DOI] [PubMed] [Google Scholar]
- Hoffman HS, Ison JR. Reflex modification in the domain of startle: I. Some empirical findings and their implications for how the nervous system processes sensory input. Psychol Rev. 1980;87(2):175–189. [PubMed] [Google Scholar]
- Honer WG, Falkai P, Bayer TA, Xie J, Hu L, Li HY, Arango V, Mann JJ, Dwork AJ, Trimble WS. Abnormalities of SNARE Mechanism Proteins in Anterior Frontal Cortex in Severe Mental Illness. Cerebral Cortex. 2002;12(4):349–356. doi: 10.1093/cercor/12.4.349. [DOI] [PubMed] [Google Scholar]
- Houston JE, Murphy J, Adamson G, Stringer M, Shevlin M. Childhood sexual abuse, early cannabis use, and psychosis: testing an interaction model based on the National Comorbidity Survey. Schizophr Bull. 2008;34(3):580–585. doi: 10.1093/schbul/sbm127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Houston JE, Murphy J, Shevlin M, Adamson G. Cannabis use and psychosis: re-visiting the role of childhood trauma. Psychol Med. 2011;41(11):2339–2348. doi: 10.1017/S0033291711000559. [DOI] [PubMed] [Google Scholar]
- Howell KR, Pillai A. Long-Term Effects of Prenatal Hypoxia on Schizophrenia-Like Phenotype in Heterozygous Reeler Mice. Mol Neurobiol. 2016;53(5):3267–3276. doi: 10.1007/s12035-015-9265-4. [DOI] [PubMed] [Google Scholar]
- 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]
- 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(1):32–37. doi: 10.1016/j.bbr.2009.08.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ishihama T, Ago Y, Shintani N, Hashimoto H, Baba A, Takuma K, Matsuda T. Environmental factors during early developmental period influence psychobehavioral abnormalities in adult PACAP-deficient mice. Behav Brain Res. 2010;209(2):274–280. doi: 10.1016/j.bbr.2010.02.009. [DOI] [PubMed] [Google Scholar]
- Jaaro-Peled H, Hayashi-Takagi A, Seshadri S, Kamiya A, Brandon NJ, Sawa A. Neurodevelopmental mechanisms of schizophrenia: understanding disturbed postnatal brain maturation through neuregulin-1-ErbB4 and DISC1. Trends in neurosciences. 2009;32(9):485–495. doi: 10.1016/j.tins.2009.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jeans AF, Oliver PL, Johnson R, Capogna M, Vikman J, Molnár Z, Babbs A, Partridge CJ, Salehi A, Bengtsson M, Eliasson L, Rorsman P, Davies KE. A dominant mutation in Snap25 causes impaired vesicle trafficking, sensorimotor gating, and ataxia in the blind-drunk mouse. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(7):2431–2436. doi: 10.1073/pnas.0610222104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ji B, Higa KK, Kim M, Zhou L, Young JW, Geyer MA, Zhou X. Inhibition of protein translation by the DISC1-Boymaw fusion gene from a Scottish family with major psychiatric disorders. Hum Mol Genet. 2014;23(21):5683–5705. doi: 10.1093/hmg/ddu285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang Z, Rompala GR, Zhang S, Cowell RM, Nakazawa K. Social isolation exacerbates schizophrenia-like phenotypes via oxidative stress in cortical interneurons. Biol Psychiatry. 2013;73(10):1024–1034. doi: 10.1016/j.biopsych.2012.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johns LC, Cannon M, Singleton N, Murray RM, Farrell M, Brugha T, Bebbington P, Jenkins R, Meltzer H. Prevalence and correlates of self-reported psychotic symptoms in the British population. Br J Psychiatry. 2004;185:298–305. doi: 10.1192/bjp.185.4.298. [DOI] [PubMed] [Google Scholar]
- Jurado S, Goswami D, Zhang Y, Miñano Molina AJ, Südhof TC, Malenka RC. LTP Requires a Unique Postsynaptic SNARE Fusion Machinery. Neuron. 2013;77(3):542–558. doi: 10.1016/j.neuron.2012.11.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kadkhodaei B, Ito T, Joodmardi E, Mattsson B, Rouillard C, Carta M, Muramatsu SI, Sumi-Ichinose C, Nomura T, Metzger D, Chambon P, Lindqvist E, Larsson NG, Olson L, Björklund A, Ichinose H, Perlmann T. Nurr1 Is Required for Maintenance of Maturing and Adult Midbrain Dopamine Neurons. The Journal of Neuroscience. 2009;29(50):15923–15932. doi: 10.1523/JNEUROSCI.3910-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kantrowitz JT, Nolan KA, Sen S, Simen AA, Lachman HM, Bowers MB., Jr Adolescent cannabis use, psychosis and catechol-O-methyltransferase genotype in African Americans and Caucasians. Psychiatr Q. 2009;80(4):213–218. doi: 10.1007/s11126-009-9108-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kedzior KK, Martin-Iverson MT. Chronic cannabis use is associated with attention-modulated reduction in prepulse inhibition of the startle reflex in healthy humans. J Psychopharmacol. 2006;20(4):471–484. doi: 10.1177/0269881105057516. [DOI] [PubMed] [Google Scholar]
- Kedzior KK, Martin-Iverson MT. Attention-dependent reduction in prepulse inhibition of the startle reflex in cannabis users and schizophrenia patients–a pilot study. Eur J Pharmacol. 2007;560(2–3):176–182. doi: 10.1016/j.ejphar.2007.01.032. [DOI] [PubMed] [Google Scholar]
- Kelly BD, O’Callaghan E, Waddington JL, Feeney L, Browne S, Scully PJ, Clarke M, Quinn JF, McTigue O, Morgan MG, Kinsella A, Larkin C. Schizophrenia and the city: A review of literature and prospective study of psychosis and urbanicity in Ireland. Schizophr Res. 2010;116(1):75–89. doi: 10.1016/j.schres.2009.10.015. [DOI] [PubMed] [Google Scholar]
- Kendler KS, Gardner CO. Dependent stressful life events and prior depressive episodes in the prediction of major depression: the problem of causal inference in psychiatric epidemiology. Arch Gen Psychiatry. 2010;67(11):1120–1127. doi: 10.1001/archgenpsychiatry.2010.136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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(6):591–596. doi: 10.1016/j.biopsych.2006.02.033. [DOI] [PubMed] [Google Scholar]
- Keshavan MS, Hogarty GE. Brain maturational processes and delayed onset in schizophrenia. Dev Psychopathol. 1999;11(3):525–543. doi: 10.1017/s0954579499002199. [DOI] [PubMed] [Google Scholar]
- Kilian S, Burns JK, Seedat S, Asmal L, Chiliza B, Du Plessis S, Olivier MR, Kidd M, Emsley R. Factors Moderating the Relationship Between Childhood Trauma and Premorbid Adjustment in First-Episode Schizophrenia. PLoS One. 2017;12(1):e0170178. doi: 10.1371/journal.pone.0170178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kjaerby C, Bundgaard C, Fejgin K, Kristiansen U, Dalby NO. Repeated potentiation of the metabotropic glutamate receptor 5 and the alpha 7 nicotinic acetylcholine receptor modulates behavioural and GABAergic deficits induced by early postnatal phencyclidine (PCP) treatment. Neuropharmacology. 2013;72:157–168. doi: 10.1016/j.neuropharm.2013.04.041. [DOI] [PubMed] [Google Scholar]
- Klug M, van den Buuse M. Chronic cannabinoid treatment during young adulthood induces sex-specific behavioural deficits in maternally separated rats. Behavioural Brain Research. 2012;233(2):305–313. doi: 10.1016/j.bbr.2012.05.019. [DOI] [PubMed] [Google Scholar]
- Klug M, van den Buuse M. An investigation into “two hit” effects of BDNF deficiency and young-adult cannabinoid receptor stimulation on prepulse inhibition regulation and memory in mice. Frontiers in behavioral neuroscience. 2013;7:149. doi: 10.3389/fnbeh.2013.00149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koenig JI. Schizophrenia: a unique translational opportunity in behavioral neuroendocrinology. Horm Behav. 2006;50(4):602–611. doi: 10.1016/j.yhbeh.2006.06.027. [DOI] [PubMed] [Google Scholar]
- 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(2):251–261. doi: 10.1016/j.bbr.2004.05.030. [DOI] [PubMed] [Google Scholar]
- Koenig JI, Kirkpatrick B, Lee P. Glucocorticoid hormones and early brain development in schizophrenia. Neuropsychopharmacology. 2002;27(2):309–318. doi: 10.1016/S0893-133X(01)00396-7. [DOI] [PubMed] [Google Scholar]
- Kohl S, Heekeren K, Klosterkötter J, Kuhn J. Prepulse inhibition in psychiatric disorders – Apart from schizophrenia. Journal of Psychiatric Research. 2013;47(4):445–452. doi: 10.1016/j.jpsychires.2012.11.018. [DOI] [PubMed] [Google Scholar]
- Konings M, Stefanis N, Kuepper R, de Graaf R, ten Have M, van Os J, Bakoula C, Henquet C. Replication in two independent population-based samples that childhood maltreatment and cannabis use synergistically impact on psychosis risk. Psychol Med. 2012;42(1):149–159. doi: 10.1017/S0033291711000973. [DOI] [PubMed] [Google Scholar]
- Kumari V, Fannon D, Geyer MA, Premkumar P, Antonova E, Simmons A, Kuipers E. Cortical grey matter volume and sensorimotor gating in schizophrenia. Cortex. 2008;44(9):1206–1214. doi: 10.1016/j.cortex.2007.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumari V, Gray JA, Geyer MA, ffytche D, Soni W, Mitterschiffthaler MT, Vythelingum GN, Simmons A, Williams SC, Sharma T. Neural correlates of tactile prepulse inhibition: a functional MRI study in normal and schizophrenic subjects. Psychiatry Res. 2003;122(2):99–113. doi: 10.1016/s0925-4927(02)00123-3. [DOI] [PubMed] [Google Scholar]
- 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]
- 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(1–2):133–144. doi: 10.1016/s0166-4328(99)00122-9. [DOI] [PubMed] [Google Scholar]
- Leon-Carrion J, Garcia-Orza J, Perez-Santamaria FJ. Development of the inhibitory component of the executive functions in children and adolescents. Int J Neurosci. 2004;114(10):1291–1311. doi: 10.1080/00207450490476066. [DOI] [PubMed] [Google Scholar]
- Lewis CM, Levinson DF, Wise LH, DeLisi LE, Straub RE, Hovatta I, Williams NM, Schwab SG, Pulver AE, Faraone SV, Brzustowicz LM, Kaufmann CA, Garver DL, Gurling HMD, Lindholm E, Coon H, Moises HW, Byerley W, Shaw SH, Mesen A, Sherrington R, O’Neill FA, Walsh D, Kendler KS, Ekelund J, Paunio T, Lönnqvist J, Peltonen L, O’Donovan MC, Owen MJ, Wildenauer DB, Maier W, Nestadt G, Blouin JL, Antonarakis SE, Mowry BJ, Silverman JM, Crowe RR, Cloninger CR, Tsuang MT, Malaspina D, Harkavy-Friedman JM, Svrakic DM, Bassett AS, Holcomb J, Kalsi G, McQuillin A, Brynjolfson J, Sigmundsson T, Petursson H, Jazin E, Zoëga T, Helgason T. Genome Scan Meta-Analysis of Schizophrenia and Bipolar Disorder, Part II: Schizophrenia. American Journal of Human Genetics. 2003;73(1):34–48. doi: 10.1086/376549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lewis DA, Levitt P. Schizophrenia as a disorder of neurodevelopment. Annu Rev Neurosci. 2002;25:409–432. doi: 10.1146/annurev.neuro.25.112701.142754. [DOI] [PubMed] [Google Scholar]
- Light G, Greenwood TA, Swerdlow NR, Calkins ME, Freedman R, Green MF, Gur RE, Gur RC, Lazzeroni LC, Nuechterlein KH, Olincy A, Radant AD, Seidman LJ, Siever LJ, Silverman JM, Sprock J, Stone WS, Sugar CA, Tsuang DW, Tsuang MT, Turetsky BI, Braff DL. Comparison of the heritability of schizophrenia and endophenotypes in the COGS-1 family study. Schizophr Bull. 2014;40(6):1404–1411. doi: 10.1093/schbul/sbu064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lim AL, Taylor DA, Malone DT. A two-hit model: behavioural investigation of the effect of combined neonatal MK-801 administration and isolation rearing in the rat. Journal of Psychopharmacology. 2012;26(9):1252–1264. doi: 10.1177/0269881111430751. [DOI] [PubMed] [Google Scholar]
- Lipina TV, Haque FN, McGirr A, Boutros PC, Berger T, Mak TW, Roder JC, Wong AH. Prophylactic valproic acid treatment prevents schizophrenia-related behaviour in Disc1-L100P mutant mice. PLoS One. 2012;7(12):e51562. doi: 10.1371/journal.pone.0051562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lipina TV, Kaidanovich-Beilin O, Patel S, Wang M, Clapcote SJ, Liu F, Woodgett JR, Roder JC. Genetic and pharmacological evidence for schizophrenia-related Disc1 interaction with GSK-3. Synapse. 2011;65(3):234–248. doi: 10.1002/syn.20839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lipina TV, Zai C, Hlousek D, Roder JC, Wong AH. Maternal immune activation during gestation interacts with Disc1 point mutation to exacerbate schizophrenia-related behaviors in mice. J Neurosci. 2013;33(18):7654–7666. doi: 10.1523/JNEUROSCI.0091-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Long LE, Chesworth R, Huang XF, McGregor IS, Arnold JC, Karl T. Transmembrane domain Nrg1 mutant mice show altered susceptibility to the neurobehavioural actions of repeated THC exposure in adolescence. Int J Neuropsychopharmacol. 2013;16(1):163–175. doi: 10.1017/S1461145711001854. [DOI] [PubMed] [Google Scholar]
- Ludewig K, Geyer MA, Vollenweider FX. Deficits in prepulse inhibition and habituation in never-medicated, first-episode schizophrenia. Biol Psychiatry. 2003;54(2):121–128. doi: 10.1016/s0006-3223(02)01925-x. [DOI] [PubMed] [Google Scholar]
- Ludewig S, Ludewig K, Geyer MA, Hell D, Vollenweider FX. Prepulse inhibition deficits in patients with panic disorder. Depress Anxiety. 2002;15(2):55–60. doi: 10.1002/da.10026. [DOI] [PubMed] [Google Scholar]
- Luthi-Carter R, Barczak AK, Speno H, Coyle JT. Hydrolysis of the neuropeptide N-acetylaspartylglutamate (NAAG) by cloned human glutamate carboxypeptidase II. Brain Res. 1998;795(1–2):341–348. doi: 10.1016/s0006-8993(98)00244-3. [DOI] [PubMed] [Google Scholar]
- Ma TM, Abazyan S, Abazyan B, Nomura J, Yang C, Seshadri S, Sawa A, Snyder SH, Pletnikov MV. Pathogenic disruption of DISC1-serine racemase binding elicits schizophrenia-like behavior via D-serine depletion. Mol Psychiatry. 2013;18(5):557–567. doi: 10.1038/mp.2012.97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mackeprang T, Kristiansen KT, Glenthoj BY. Effects of antipsychotics on prepulse inhibition of the startle response in drug-naive schizophrenic patients. Biol Psychiatry. 2002;52(9):863–873. doi: 10.1016/s0006-3223(02)01409-9. [DOI] [PubMed] [Google Scholar]
- Manning EE, van den Buuse M. BDNF deficiency and young-adult methamphetamine induce sex-specific effects on prepulse inhibition regulation. Front Cell Neurosci. 2013;7:92. doi: 10.3389/fncel.2013.00092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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(4):871–879. doi: 10.1017/s0033291798006898. [DOI] [PubMed] [Google Scholar]
- Marriott AL, Tasker RA, Ryan CL, Doucette TA. Alterations to prepulse inhibition magnitude and latency in adult rats following neonatal treatment with domoic acid and social isolation rearing. Behavioural Brain Research. 2016;298(Part B):310–317. doi: 10.1016/j.bbr.2015.11.009. [DOI] [PubMed] [Google Scholar]
- Mason O, Startup M, Halpin S, Schall U, Conrad A, Carr V. Risk factors for transition to first episode psychosis among individuals with ‘at-risk mental states’. Schizophr Res. 2004;71(2–3):227–237. doi: 10.1016/j.schres.2004.04.006. [DOI] [PubMed] [Google Scholar]
- McAlonan GM, Daly E, Kumari V, Critchley HD, van Amelsvoort T, Suckling J, Simmons A, Sigmundsson T, Greenwood K, Russell A, Schmitz N, Happe F, Howlin P, Murphy DG. Brain anatomy and sensorimotor gating in Asperger’s syndrome. Brain. 2002;125(Pt 7):1594–1606. doi: 10.1093/brain/awf150. [DOI] [PubMed] [Google Scholar]
- McCarroll SA, Feng G, Hyman SE. Genome-scale neurogenetics: methodology and meaning. Nat Neurosci. 2014;17(6):756–763. doi: 10.1038/nn.3716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McKetin R, Lubman DI, Baker AL, Dawe S, Ali RL. Dose-related psychotic symptoms in chronic methamphetamine users: Evidence from a prospective longitudinal study. JAMA Psychiatry. 2013;70(3):319–324. doi: 10.1001/jamapsychiatry.2013.283. [DOI] [PubMed] [Google Scholar]
- Mednick SA, Machon RA, Huttunen MO, Bonett D. Adult schizophrenia following prenatal exposure to an influenza epidemic. Arch Gen Psychiatry. 1988;45(2):189–192. doi: 10.1001/archpsyc.1988.01800260109013. [DOI] [PubMed] [Google Scholar]
- Meyer U. Prenatal poly(i:C) exposure and other developmental immune activation models in rodent systems. Biol Psychiatry. 2014;75(4):307–315. doi: 10.1016/j.biopsych.2013.07.011. [DOI] [PubMed] [Google Scholar]
- Meyer U, Feldon J. Epidemiology-driven neurodevelopmental animal models of schizophrenia. Prog Neurobiol. 2009a;90(3):285–326. doi: 10.1016/j.pneurobio.2009.10.018. [DOI] [PubMed] [Google Scholar]
- Meyer U, Feldon J. Neural basis of psychosis-related behaviour in the infection model of schizophrenia. Behav Brain Res. 2009b;204(2):322–334. doi: 10.1016/j.bbr.2008.12.022. [DOI] [PubMed] [Google Scholar]
- 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. The Journal of Neuroscience. 2006;26(18):4752–4762. doi: 10.1523/JNEUROSCI.0099-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. 2010;36(3):607–623. doi: 10.1093/schbul/sbn131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Millan MJ, Andrieux A, Bartzokis G, Cadenhead K, Dazzan P, Fusar-Poli P, Gallinat J, Giedd J, Grayson DR, Heinrichs M, Kahn R, Krebs MO, Leboyer M, Lewis D, Marin O, Marin P, Meyer-Lindenberg A, McGorry P, McGuire P, Owen MJ, Patterson P, Sawa A, Spedding M, Uhlhaas P, Vaccarino F, Wahlestedt C, Weinberger D. Altering the course of schizophrenia: progress and perspectives. Nat Rev Drug Discov. 2016;15(7):485–515. doi: 10.1038/nrd.2016.28. [DOI] [PubMed] [Google Scholar]
- Millar JK, Wilson-Annan JC, Anderson S, Christie S, Taylor MS, Semple CA, Devon RS, St Clair DM, Muir WJ, Blackwood DH, Porteous DJ. Disruption of two novel genes by a translocation co-segregating with schizophrenia. Hum Mol Genet. 2000;9(9):1415–1423. doi: 10.1093/hmg/9.9.1415. [DOI] [PubMed] [Google Scholar]
- Miller P, Lawrie SM, Hodges A, Clafferty R, Cosway R, Johnstone EC. Genetic liability, illicit drug use, life stress and psychotic symptoms: preliminary findings from the Edinburgh study of people at high risk for schizophrenia. Soc Psychiatry Psychiatr Epidemiol. 2001;36(7):338–342. doi: 10.1007/s001270170038. [DOI] [PubMed] [Google Scholar]
- 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(4):346–354. doi: 10.1111/j.1601-183X.2005.00172.x. [DOI] [PubMed] [Google Scholar]
- Moller P, Husby R. The Initial Prodrome in Schizophrenia: Searching for Naturalistic Core Dimensions of Experience and Behavior. Schizophr Bull. 2000;26(1):217–232. doi: 10.1093/oxfordjournals.schbul.a033442. [DOI] [PubMed] [Google Scholar]
- Moore H. The role of rodent models in the discovery of new treatments for schizophrenia: updating our strategy. Schizophr Bull. 2010;36(6):1066–1072. doi: 10.1093/schbul/sbq106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moore TH, Zammit S, Lingford-Hughes A, Barnes TR, Jones PB, Burke M, Lewis G. Cannabis use and risk of psychotic or affective mental health outcomes: a systematic review. Lancet. 2007;370(9584):319–328. doi: 10.1016/S0140-6736(07)61162-3. [DOI] [PubMed] [Google Scholar]
- Moran P, Stokes J, Marr J, Bock G, Desbonnet L, Waddington J, Tuathaigh C. Gene × Environment Interactions in Schizophrenia: Evidence from Genetic Mouse Models. Neural Plasticity. 2016;2016:23. doi: 10.1155/2016/2173748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mortensen PB, Pedersen CB, Westergaard T, Wohlfahrt J, Ewald H, Mors O, Andersen PK, Melbye M. Effects of Family History and Place and Season of Birth on the Risk of Schizophrenia. New England Journal of Medicine. 1999;340(8):603–608. doi: 10.1056/NEJM199902253400803. [DOI] [PubMed] [Google Scholar]
- Murphy J, Houston JE, Shevlin M, Adamson G. Childhood sexual trauma, cannabis use and psychosis: statistically controlling for pre-trauma psychosis and psychopathology. Soc Psychiatry Psychiatr Epidemiol. 2013;48(6):853–861. doi: 10.1007/s00127-012-0592-8. [DOI] [PubMed] [Google Scholar]
- Murray RM, McDonald C, Bramon E. Neurodevelopmental impairment, dopamine sensitisation, and social adversity in schizophrenia. World Psychiatry. 2002;1(3):137–145. [PMC free article] [PubMed] [Google Scholar]
- Need AC, Goldstein DB. Schizophrenia genetics comes of age. Neuron. 2014;83(4):760–763. doi: 10.1016/j.neuron.2014.08.015. [DOI] [PubMed] [Google Scholar]
- Neuner I, Stocker T, Kellermann T, Ermer V, Wegener HP, Eickhoff SB, Schneider F, Shah NJ. Electrophysiology meets fMRI: neural correlates of the startle reflex assessed by simultaneous EMG-fMRI data acquisition. Hum Brain Mapp. 2010;31(11):1675–1685. doi: 10.1002/hbm.20965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nielsen SM, Toftdahl NG, Nordentoft M, Hjorthoj C. Association between alcohol, cannabis, and other illicit substance abuse and risk of developing schizophrenia: a nationwide population based register study. Psychol Med. 2017:1–10. doi: 10.1017/S0033291717000162. [DOI] [PubMed] [Google Scholar]
- Nusbaum MP, Contreras D. Sensorimotor gating: startle submits to presynaptic inhibition. Curr Biol. 2004;14(6):R247–249. doi: 10.1016/j.cub.2004.02.059. [DOI] [PubMed] [Google Scholar]
- O’Callaghan E, Sham P, Takei N, Glover G, Murray RM. Schizophrenia after prenatal exposure to 1957 A2 influenza epidemic. Lancet. 1991;337(8752):1248–1250. doi: 10.1016/0140-6736(91)92919-s. [DOI] [PubMed] [Google Scholar]
- O’Leary C, Desbonnet L, Clarke N, Petit E, Tighe O, Lai D, Harvey R, Waddington JL, O’Tuathaigh C. Phenotypic effects of maternal immune activation and early postnatal milieu in mice mutant for the schizophrenia risk gene neuregulin-1. Neuroscience. 2014;277:294–305. doi: 10.1016/j.neuroscience.2014.06.028. [DOI] [PubMed] [Google Scholar]
- O’Tuathaigh CM, Waddington JL. Closing the translational gap between mutant mouse models and the clinical reality of psychotic illness. Neurosci Biobehav Rev. 2015;58:19–35. doi: 10.1016/j.neubiorev.2015.01.016. [DOI] [PubMed] [Google Scholar]
- Oliver PL, Davies KE. Interaction between environmental and genetic factors modulates schizophrenic endophenotypes in the Snap-25 mouse mutant blind-drunk. Human Molecular Genetics. 2009;18(23):4576–4589. doi: 10.1093/hmg/ddp425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Opler MGA, Brown AS, Graziano J, Desai M, Zheng W, Schaefer C, Factor-Litvak P, Susser ES. Prenatal lead exposure, delta-aminolevulinic acid, and schizophrenia. Environmental Health Perspectives. 2004;112(5):548–552. doi: 10.1289/ehp.6777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ornitz EM, Hanna GL, de Traversay J. Prestimulation-induced startle modulation in attention-deficit hyperactivity disorder and nocturnal enuresis. Psychophysiology. 1992;29(4):437–451. doi: 10.1111/j.1469-8986.1992.tb01717.x. [DOI] [PubMed] [Google Scholar]
- Owen MJ, Sawa A, Mortensen PB. Schizophrenia. Lancet. 2016;388(10039):86–97. doi: 10.1016/S0140-6736(15)01121-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 Research. 2004;996(2):193–201. doi: 10.1016/j.brainres.2003.09.077. [DOI] [PubMed] [Google Scholar]
- Pantelis C, Pantelis C, Yücel M, Wood SJ, McGorry PD, Velakoulis D. Early and Late Neurodevelopmental Disturbances in Schizophrenia and Their Functional Consequences. Australian & New Zealand Journal of Psychiatry. 2003;37(4):399–406. doi: 10.1046/j.1440-1614.2003.01193.x. [DOI] [PubMed] [Google Scholar]
- Patterson PH. Immune involvement in schizophrenia and autism: etiology, pathology and animal models. Behav Brain Res. 2009;204(2):313–321. doi: 10.1016/j.bbr.2008.12.016. [DOI] [PubMed] [Google Scholar]
- Perry W, Minassian A, Feifel D, Braff DL. Sensorimotor gating deficits in bipolar disorder patients with acute psychotic mania. Biol Psychiatry. 2001;50(6):418–424. doi: 10.1016/s0006-3223(01)01184-2. [DOI] [PubMed] [Google Scholar]
- Perry W, Minassian A, Lopez B, Maron L, Lincoln A. Sensorimotor gating deficits in adults with autism. Biol Psychiatry. 2007;61(4):482–486. doi: 10.1016/j.biopsych.2005.09.025. [DOI] [PubMed] [Google Scholar]
- Petrovsky N, Quednow BB, Ettinger U, Schmechtig A, Mossner R, Collier DA, Kuhn KU, Maier W, Wagner M, Kumari V. Sensorimotor gating is associated with CHRNA3 polymorphisms in schizophrenia and healthy volunteers. Neuropsychopharmacology. 2010;35(7):1429–1439. doi: 10.1038/npp.2010.12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Powell SB. Models of neurodevelopmental abnormalities in schizophrenia. Curr Top Behav Neurosci. 2010;4:435–481. doi: 10.1007/7854_2010_57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Powell SB, Swerdlow NR. Social isolation rearing and sensorimotor gating in rat models of relevance to schizophrenia: What we know, and what we don’t. In: Pletnikov M, Waddington JL, editors. Modeling Psychopathological Dimensions of Schizophrenia. Elsevier; 2015. [Google Scholar]
- Powell SB, Weber M, Geyer MA. Genetic models of sensorimotor gating: relevance to neuropsychiatric disorders. Curr Top Behav Neurosci. 2012;12:251–318. doi: 10.1007/7854_2011_195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Powell SB, Zhou X, Geyer MA. Prepulse inhibition and genetic mouse models of schizophrenia. Behav Brain Res. 2009 doi: 10.1016/j.bbr.2009.04.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Quednow BB, Kuhn KU, Hoenig K, Maier W, Wagner M. Prepulse inhibition and habituation of acoustic startle response in male MDMA (‘ecstasy’) users, cannabis users, and healthy controls. Neuropsychopharmacology. 2004;29(5):982–990. doi: 10.1038/sj.npp.1300396. [DOI] [PubMed] [Google Scholar]
- Quednow BB, Wagner M, Mossner R, Maier W, Kuhn KU. Sensorimotor Gating of Schizophrenia Patients Depends on Catechol O-Methyltransferase Val158Met Polymorphism. Schizophr Bull. 2008;36(2):341–346. doi: 10.1093/schbul/sbn088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Quesseveur G, David DJ, Gaillard MC, Pla P, Wu MV, Nguyen HT, Nicolas V, Auregan G, David I, Dranovsky A, Hantraye P, Hen R, Gardier AM, Deglon N, Guiard BP. BDNF overexpression in mouse hippocampal astrocytes promotes local neurogenesis and elicits anxiolytic-like activities. Transl Psychiatry. 2013;3:e253. doi: 10.1038/tp.2013.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ramsay H, Kelleher I, Flannery P, Clarke MC, Lynch F, Harley M, Connor D, Fitzpatrick C, Morris DW, Cannon M. Relationship between the COMT-Val158Met and BDNF-Val66Met polymorphisms, childhood trauma and psychotic experiences in an adolescent general population sample. PLoS One. 2013;8(11):e79741. doi: 10.1371/journal.pone.0079741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rapoport JL, Addington AM, Frangou S, Psych MRC. The neurodevelopmental model of schizophrenia: update 2005. Mol Psychiatry. 2005;10(5):434–449. doi: 10.1038/sj.mp.4001642. [DOI] [PubMed] [Google Scholar]
- Rapoport JL, Giedd JN, Gogtay N. Neurodevelopmental model of schizophrenia: update 2012. Mol Psychiatry. 2012;17(12):1228–1238. doi: 10.1038/mp.2012.23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Read J, Bentall RP. Negative childhood experiences and mental health: theoretical, clinical and primary prevention implications. Br J Psychiatry. 2012;200(2):89–91. doi: 10.1192/bjp.bp.111.096727. [DOI] [PubMed] [Google Scholar]
- Richetto J, Feldon J, Riva MA, Meyer U. Comparison of the long-term consequences of withdrawal from repeated amphetamine exposure in adolescence and adulthood on information processing and locomotor sensitization in mice. European Neuropsychopharmacology. 2013;23(2):160–170. doi: 10.1016/j.euroneuro.2012.04.005. [DOI] [PubMed] [Google Scholar]
- Rojas P, Joodmardi E, Hong Y, Perlmann T, Ogren SO. Adult mice with reduced Nurr1 expression: an animal model for schizophrenia. Mol Psychiatry. 2007;12(8):756–766. doi: 10.1038/sj.mp.4001993. [DOI] [PubMed] [Google Scholar]
- Rose PK, Scott SH. Sensory-motor control: a long-awaited behavioral correlate of presynaptic inhibition. Nat Neurosci. 2003;6(12):1243–1245. doi: 10.1038/nn1203-1243. [DOI] [PubMed] [Google Scholar]
- Ross CA, Margolis RL, Reading SA, Pletnikov M, Coyle JT. Neurobiology of schizophrenia. Neuron. 2006;52:139–153. doi: 10.1016/j.neuron.2006.09.015. [DOI] [PubMed] [Google Scholar]
- Roussos P, Giakoumaki SG, Adamaki E, Bitsios P. The influence of schizophrenia-related neuregulin-1 polymorphisms on sensorimotor gating in healthy males. Biol Psychiatry. 2011;69(5):479–486. doi: 10.1016/j.biopsych.2010.09.009. [DOI] [PubMed] [Google Scholar]
- Roussos P, Giakoumaki SG, Rogdaki M, Pavlakis S, Frangou S, Bitsios P. Prepulse inhibition of the startle reflex depends on the catechol O-methyltransferase Val158Met gene polymorphism. Psychol Med. 2008:1–8. doi: 10.1017/S0033291708002912. [DOI] [PubMed] [Google Scholar]
- 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(4):767–774. doi: 10.1016/j.pbb.2004.01.017. [DOI] [PubMed] [Google Scholar]
- Schaevitz LR, Picker JD, Rana J, Kolodny NH, Shane B, Berger-Sweeney JE, Coyle JT. Glutamate carboxypeptidase II and folate deficiencies result in reciprocal protection against cognitive and social deficits in mice: implications for neurodevelopmental disorders. Dev Neurobiol. 2012;72(6):891–905. doi: 10.1002/dneu.21000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schneider M, Drews E, Koch M. Behavioral effects in adult rats of chronic prepubertal treatment with the cannabinoid receptor agonist WIN 55,212-2. Behavioural pharmacology. 2005;16(5–6):447–454. doi: 10.1097/00008877-200509000-00018. [DOI] [PubMed] [Google Scholar]
- Schneider M, Koch M. The cannabinoid agonist WIN 55,212-2 reduces sensorimotor gating and recognition memory in rats. Behavioural pharmacology. 2002;13(1):29–37. doi: 10.1097/00008877-200202000-00003. [DOI] [PubMed] [Google Scholar]
- Schneider M, Koch M. Chronic pubertal, but not adult chronic cannabinoid treatment impairs sensorimotor gating, recognition memory, and the performance in a progressive ratio task in adult rats. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology. 2003;28(10):1760–1769. doi: 10.1038/sj.npp.1300225. [DOI] [PubMed] [Google Scholar]
- Schoenrock SA, Tarantino LM. Developmental vitamin D deficiency and schizophrenia: the role of animal models. Genes Brain Behav. 2016;15(1):45–61. doi: 10.1111/gbb.12271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scholes KE, Martin-Iverson MT. Alterations to pre-pulse inhibition (PPI) in chronic cannabis users are secondary to sustained attention deficits. Psychopharmacology (Berl) 2009;207(3):469–484. doi: 10.1007/s00213-009-1679-0. [DOI] [PubMed] [Google Scholar]
- Schroeder A, Buret L, Hill RA, van den Buuse M. Gene-environment interaction of reelin and stress in cognitive behaviours in mice: Implications for schizophrenia. Behav Brain Res. 2015;287:304–314. doi: 10.1016/j.bbr.2015.03.063. [DOI] [PubMed] [Google Scholar]
- Schubert CR, Xi HS, Wendland JR, O’Donnell P. Translating human genetics into novel treatment targets for schizophrenia. Neuron. 2014;84(3):537–541. doi: 10.1016/j.neuron.2014.10.037. [DOI] [PubMed] [Google Scholar]
- 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. 1999;38(2–3):85–91. doi: 10.1016/s0920-9964(99)00005-5. [DOI] [PubMed] [Google Scholar]
- Seshadri S, Kamiya A, Yokota Y, Prikulis I, Kano S-i, Hayashi-Takagi A, Stanco A, Eom TY, Rao S, Ishizuka K, Wong P, Korth C, Anton ES, Sawa A. Disrupted-in-Schizophrenia-1 expression is regulated by β-site amyloid precursor protein cleaving enzyme-1–neuregulin cascade. Proceedings of the National Academy of Sciences. 2010;107(12):5622–5627. doi: 10.1073/pnas.0909284107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shevlin M, Houston JE, Dorahy MJ, Adamson G. Cumulative traumas and psychosis: an analysis of the national comorbidity survey and the British Psychiatric Morbidity Survey. Schizophr Bull. 2008;34(1):193–199. doi: 10.1093/schbul/sbm069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi L, Fatemi SH, Sidwell RW, Patterson PH. Maternal influenza infection causes marked behavioral and pharmacological changes in the offspring. J Neurosci. 2003;23(1):297–302. doi: 10.1523/JNEUROSCI.23-01-00297.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith SEP, Li J, Garbett K, Mirnics K, Patterson PH. Maternal Immune Activation Alters Fetal Brain Development through Interleukin-6. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2007;27(40):10695–10702. doi: 10.1523/JNEUROSCI.2178-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sobin C, Kiley-Brabeck K, Karayiorgou M. Lower prepulse inhibition in children with the 22q11 deletion syndrome. Am J Psychiatry. 2005;162(6):1090–1099. doi: 10.1176/appi.ajp.162.6.1090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sorensen HJ, Mortensen EL, Reinisch JM, Mednick SA. Association between prenatal exposure to bacterial infection and risk of schizophrenia. Schizophr Bull. 2009;35(3):631–637. doi: 10.1093/schbul/sbn121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Strauss L, Brink CB, Möller M, Stein DJ, Harvey BH. Late-Life Effects of Chronic Methamphetamine Exposure during Puberty on Behaviour and Corticostriatal Mono-Amines in Social Isolation-Reared Rats. Developmental Neuroscience. 2014;36(1):18–28. doi: 10.1159/000357495. [DOI] [PubMed] [Google Scholar]
- Sun XL, Chen BY, Duan L, Xia Y, Luo ZJ, Wang JJ, Rao ZR, Chen LW. The proform of glia cell line-derived neurotrophic factor: a potentially biologically active protein. Mol Neurobiol. 2014;49(1):234–250. doi: 10.1007/s12035-013-8515-6. [DOI] [PubMed] [Google Scholar]
- 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(1):25–31. doi: 10.1001/archpsyc.1996.01830010027005. [DOI] [PubMed] [Google Scholar]
- Swerdlow NR, Benbow CH, Zisook S, Geyer MA, Braff DL. A preliminary assessment of sensorimotor gating in patients with obsessive compulsive disorder. Biol Psychiatry. 1993;33(4):298–301. doi: 10.1016/0006-3223(93)90300-3. [DOI] [PubMed] [Google Scholar]
- Swerdlow NR, Braff DL, Geyer MA. Sensorimotor gating of the startle reflex: what we said 25 years ago, what has happened since then, and what comes next. Journal of Psychopharmacology. 2016;30(11):1072–1081. doi: 10.1177/0269881116661075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Swerdlow NR, Geyer MA, Braff DL. Neural circuit regulation of prepulse inhibition of startle in the rat: current knowledge and future challenges. Psychopharmacology (Berl) 2001a;156(2–3):194–215. doi: 10.1007/s002130100799. [DOI] [PubMed] [Google Scholar]
- Swerdlow NR, Karban B, Ploum Y, Sharp R, Geyer MA, Eastvold A. Tactile prepuff inhibition of startle in children with Tourette’s syndrome: in search of an “fMRI-friendly” startle paradigm. Biol Psychiatry. 2001b;50(8):578–585. doi: 10.1016/s0006-3223(01)01164-7. [DOI] [PubMed] [Google Scholar]
- Swerdlow NR, Light GA. Animal Models of Deficient Sensorimotor Gating in Schizophrenia: Are They Still Relevant? In: Robbins TW, Sahakian BJ, editors. Translational Neuropsychopharmacology. Springer International Publishing; Cham: 2016. pp. 305–325. [DOI] [PubMed] [Google Scholar]
- Swerdlow NR, Light GA, Sprock J, Calkins ME, Green MF, Greenwood TA, Gur RE, Gur RC, Lazzeroni LC, Nuechterlein KH, Radant AD, Ray A, Seidman LJ, Siever LJ, Silverman JM, Stone WS, Sugar CA, Tsuang DW, Tsuang MT, Turetsky BI, Braff DL. Deficient prepulse inhibition in schizophrenia detected by the multi-site COGS. Schizophrenia Research. 2014;152(2–3):503–512. doi: 10.1016/j.schres.2013.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Swerdlow NR, Paulsen J, Braff DL, Butters N, Geyer MA, Swenson MR. Impaired prepulse inhibition of acoustic and tactile startle response in patients with Huntington’s disease. J Neurol Neurosurg Psychiatry. 1995;58(2):192–200. doi: 10.1136/jnnp.58.2.192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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(3):331–388. doi: 10.1007/s00213-008-1072-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tanaka K, Shintani N, Hashimoto H, Kawagishi N, Ago Y, Matsuda T, Hashimoto R, Kunugi H, Yamamoto A, Kawaguchi C, Shimada T, Baba A. Psychostimulant-induced attenuation of hyperactivity and prepulse inhibition deficits in Adcyap1-deficient mice. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2006;26(19):5091–5097. doi: 10.1523/JNEUROSCI.4376-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Turetsky BI, Calkins ME, Light GA, Olincy A, Radant AD, Swerdlow NR. Neurophysiological endophenotypes of schizophrenia: the viability of selected candidate measures. Schizophr Bull. 2007;33(1):69–94. doi: 10.1093/schbul/sbl060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uher R. Gene-Environment Interactions in Severe Mental Illness. Frontiers in Psychiatry. 2014;5:48. doi: 10.3389/fpsyt.2014.00048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Valls-Sole J, Munoz JE, Valldeoriola F. Abnormalities of prepulse inhibition do not depend on blink reflex excitability: a study in Parkinson’s disease and Huntington’s disease. Clin Neurophysiol. 2004;115(7):1527–1536. doi: 10.1016/j.clinph.2004.02.014. [DOI] [PubMed] [Google Scholar]
- van den Buuse M. Modeling the Positive Symptoms of Schizophrenia in Genetically Modified Mice: Pharmacology and Methodology Aspects. Schizophrenia Bulletin. 2010;36(2):246–270. doi: 10.1093/schbul/sbp132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Os J, Bak M, Hanssen M, Bijl RV, de Graaf R, Verdoux H. Cannabis Use and Psychosis: A Longitudinal Population-based Study. American Journal of Epidemiology. 2002;156(4):319–327. doi: 10.1093/aje/kwf043. [DOI] [PubMed] [Google Scholar]
- van Rijn S, Swaab H, Magnee M, van Engeland H, Kemner C. Psychophysiological markers of vulnerability to psychopathology in men with an extra × chromosome (XXY) PLoS One. 2011;6(5):e20292. doi: 10.1371/journal.pone.0020292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Winkel R. Family-based analysis of genetic variation underlying psychosis-inducing effects of cannabis: sibling analysis and proband follow-up. Arch Gen Psychiatry. 2011;68(2):148–157. doi: 10.1001/archgenpsychiatry.2010.152. [DOI] [PubMed] [Google Scholar]
- van Winkel R, Stefanis NC, Myin-Germeys I. Psychosocial stress and psychosis. A review of the neurobiological mechanisms and the evidence for gene-stress interaction. Schizophr Bull. 2008;34(6):1095–1105. doi: 10.1093/schbul/sbn101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vaucher J, Keating BJ, Lasserre AM, Gan W, Lyall DM, Ward J, Smith DJ, Pell JP, Sattar N, Pare G, Holmes MV. Cannabis use and risk of schizophrenia: a Mendelian randomization study. Mol Psychiatry. 2017 doi: 10.1038/mp.2016.252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vinkers CH, Van Gastel WA, Schubart CD, Van Eijk KR, Luykx JJ, Van Winkel R, Joels M, Ophoff RA, Boks MP, Bruggeman R, Cahn W, de Haan L, Kahn RS, Meijer CJ, Myin-Germeys I, van Os J, Wiersma D. The effect of childhood maltreatment and cannabis use on adult psychotic symptoms is modified by the COMT Val(1)(5)(8)Met polymorphism. Schizophr Res. 2013;150(1):303–311. doi: 10.1016/j.schres.2013.07.020. [DOI] [PubMed] [Google Scholar]
- Vuillermot S, Joodmardi E, Perlmann T, Ove Ögren S, Feldon J, Meyer U. Prenatal Immune Activation Interacts with Genetic <em>Nurr1</em> Deficiency in the Development of Attentional Impairments. The Journal of Neuroscience. 2012;32(2):436–451. doi: 10.1523/JNEUROSCI.4831-11.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Waschek JA. VIP and PACAP: neuropeptide modulators of CNS inflammation, injury, and repair. British journal of pharmacology. 2013;169(3):512–523. doi: 10.1111/bph.12181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wegener N, Koch M. Behavioural disturbances and altered Fos protein expression in adult rats after chronic pubertal cannabinoid treatment. Brain research. 2009;1253:81–91. doi: 10.1016/j.brainres.2008.11.081. [DOI] [PubMed] [Google Scholar]
- 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(1–2):207–218. doi: 10.1016/s0166-4328(01)00166-8. [DOI] [PubMed] [Google Scholar]
- Wiles NJ, Zammit S, Bebbington P, Singleton N, Meltzer H, Lewis G. Self-reported psychotic symptoms in the general population: results from the longitudinal study of the British National Psychiatric Morbidity Survey. Br J Psychiatry. 2006;188:519–526. doi: 10.1192/bjp.bp.105.012179. [DOI] [PubMed] [Google Scholar]
- Xu MQ, Sun WS, Liu BX, Feng GY, Yu L, Yang L, He G, Sham P, Susser E, Clair D, He L. Prenatal Malnutrition and Adult Schizophrenia: Further Evidence From the 1959–1961 Chinese Famine. Schizophr Bull. 2009;35(3):568–576. doi: 10.1093/schbul/sbn168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yee N, Ribic A, de Roo CC, Fuchs E. Differential effects of maternal immune activation and juvenile stress on anxiety-like behaviour and physiology in adult rats: no evidence for the “double-hit hypothesis”. Behav Brain Res. 2011;224(1):180–188. doi: 10.1016/j.bbr.2011.05.040. [DOI] [PubMed] [Google Scholar]
- Zammit S, Owen MJ, Evans J, Heron J, Lewis G. Cannabis, COMT and psychotic experiences. Br J Psychiatry. 2011;199(5):380–385. doi: 10.1192/bjp.bp.111.091421. [DOI] [PubMed] [Google Scholar]
- Zammit S, Spurlock G, Williams H, Norton N, Williams N, O’Donovan MC, Owen MJ. Genotype effects of CHRNA7, CNR1 and COMT in schizophrenia: interactions with tobacco and cannabis use. Br J Psychiatry. 2007;191:402–407. doi: 10.1192/bjp.bp.107.036129. [DOI] [PubMed] [Google Scholar]
- Zornberg GL, Buka SL, Tsuang MT. The problem of obstetrical complications and schizophrenia. Schizophr Bull. 2000;26(2):249–256. doi: 10.1093/oxfordjournals.schbul.a033449. [DOI] [PubMed] [Google Scholar]