Abstract
Schizophrenia is a heterogeneous psychiatric disorder that is poorly treated with current therapies. In this brief review, we provide an update regarding the use of animal models to study schizophrenia in an attempt to understand its aetiology and develop novel therapeutic strategies. Tremendous progress has been made developing and validating rodent models that replicate the aetiologies, brain pathologies, and behavioural abnormalities associated with schizophrenia in humans. Here, models are grouped into 3 categories—developmental, drug induced, and genetic—to reflect the heterogeneous risk factors associated with schizophrenia. Each of these models is associated with varied but overlapping pathophysiology, endophenotypes, behavioural abnormalities, and cognitive impairments. Studying schizophrenia using multiple models will permit an understanding of the core features of the disease, thereby facilitating preclinical research aimed at the development and validation of better pharmacotherapies to alter the progression of schizophrenia or alleviate its debilitating symptoms.
Keywords: animal models, schizophrenia, dopamine, glutamate, 5-hydroxytryptamine (5-HT, serotonin)
Abstract
La schizophrénie est un trouble psychiatrique hétérogène qui n’est pas bien traité par les thérapies actuelles. Dans cette brève revue, nous offrons une mise à jour concernant l’utilisation de modèles animaux pour étudier la schizophrénie en vue d’en comprendre l’étiologie et d’élaborer de nouvelles stratégies thérapeutiques. Des progrès formidables ont été réalisés dans le développement et la validation de modèles rongeurs qui reproduisent les étiologies, les pathologies cérébrales et les anomalies comportementales associées à la schizophrénie chez les humains. Ici, les modèles sont groupés en 3 catégories: développementale, induite par des médicaments, et génétique pour refléter les facteurs de risque hétérogènes associés à la schizophrénie. Chacun de ces modèles est associé à une pathophysiologie, des endophénotypes, des anomalies comportementales, et des déficiences cognitives variés mais se chevauchant. Étudier la schizophrénie à l’aide de modèles multiples nous permettra de comprendre les caractéristiques essentielles de la maladie, facilitant ainsi la recherche préclinique qui vise le développement et la validation de meilleures pharmacothérapies afin de modifier la progression de la schizophrénie ou d’en atténuer les symptômes invalidants.
Schizophrenia is a severe mental illness with a lifetime prevalence of approximately 0.4% worldwide.1 It can have devastating effects on patients and their caretakers and imposes enormous costs on health care systems. Schizophrenia is a heterogeneous disorder with positive symptoms (delusions, hallucinations, thought disorders), negative symptoms (anhedonia, avolition, social withdrawal, poverty of thought), and cognitive dysfunction. Although numerous antipsychotic drugs are available to mitigate the symptoms of schizophrenia, the response rate to these drugs is lower than desired, they are slow-acting, and they often produce serious adverse side effects. While the aetiology of schizophrenia is still poorly understood and the biochemical focus has been on dopamine and glutamate, multiple neurotransmitters and neuromodulators, including 5-hydroxytryptamine (5-HT), gamma-aminobutyric acid (GABA), glycine, D-serine, and neuroactive steroids, have been implicated in its pathophysiology.2–4 Based on the limited understanding of the biological origins of schizophrenia, there is a continued need for improved animal models of the disorder to better identify the origins of the varied symptoms and to develop and validate novel therapies. This article provides a brief overview of the models currently available and the complexities involved in attempting to develop and use such models.
In general, animal models of complex psychiatric disorders are valuable for studying the neurobiological bases of these disorders and for identifying new drug targets to provide more effective treatment in the future. Preclinical models allow for more rapid monitoring of disease progression than is possible in humans, permit invasive studies of structural and molecular changes across development of the disorder, and allow for relatively easy testing of potential therapeutic agents.5 However, neuropsychiatric disorders, including schizophrenia, include symptoms such as paranoid delusions and auditory hallucinations that are uniquely human and make interpretation of results obtained from animal models more difficult.6 For example, increased locomotor activity in response to psychotomimetic compounds such as amphetamine or noncompetitive N-methyl-D-aspartate (NMDA) glutamate receptor antagonists is commonly used as an indication of positive symptoms in schizophrenia, a deficit in prepulse inhibition (PPI, where a weaker sound [prepulse] delivered prior to a stronger sound [pulse] inhibits the natural startle response to the second stimulus) is used as a proxy for sensorimotor gating problems, and changes in social interaction are used as an indicator of negative symptoms.5 While these tests provide relatively high-throughput evaluation of symptom progression or alleviation with treatment, their relationship to human symptoms can be questioned. More recently, the cognitive symptoms of the disorder have become a focus for research, an important consideration given the lack of effective treatments for these symptoms. The MATRICS initiative of the National Institute of Mental Health (NIMH) identified 7 cognitive domains specifically relevant for schizophrenia, and rodent tests have been proposed for each of these domains.7 The use of touchscreen-equipped operant conditioning chambers for studying cognition relevant to schizophrenia in rodents is increasingly common given the high face validity, ease of implementation, and high level of automation possible with these tasks.8–10 Touchscreen-equipped chambers include a touch-sensitive screen on one wall of the chamber that replaces the levers used in traditional operant conditioning tasks. During the tasks, rodents are required to touch specific areas of the screen according to a given rule to receive the food reward. The NIMH has also proposed the Research Domain Criteria (RDoC) approach for research into the biological underpinnings of behaviour, ultimately including psychopathology and mental disorders.11,12 RDoC constructs have initially been divided into 5 domains (negative valence systems, positive valence systems, cognitive systems, social processes, and arousal and regulatory systems), all of which are affected in schizophrenia.13 Work integrating animal research related to mental illness, including schizophrenia, into the RDoC framework is ongoing.14,15
As discussed in the editorial accompanying this issue, animal models are often sought that satisfy face, predictive, and construct validity. Most of the animal models currently used to study schizophrenia are developmental, drug induced, or genetic.5 The purpose of this article is to introduce readers of the Canadian Journal of Psychiatry to these strategies for studying schizophrenia in animals with a focus on recent findings. For an in-depth consideration of particular models, comprehensive articles regarding the various animal models of schizophrenia are available.5,16–37 Table 1 summarizes common phenotypes associated with animal models of schizophrenia.
Table 1.
Category | Measure | Description |
---|---|---|
Behavioural | Locomotor activity (positive symptoms) | Vigor of movement in response to a novel environment, stress, or pharmacological challenge (dopamine enhancers, NMDA receptor antagonists) is quantified. |
Sucrose preference (negative symptoms) | Preference for sweetened water over unsweetened water is measured. | |
Social interaction (negative and cognitive symptoms) | Time spent in close proximity or direct contact with an unfamiliar or less familiar animal. | |
Latent inhibition (attentional processes, cognitive symptoms) | A classical conditioning procedure whereby previously unreinforced stimuli are slower (less efficient) in generating a conditioned response than novel stimuli. | |
Memory (cognitive symptoms) | Many tests have been validated to measure various aspects of working memory, declarative memory, recognition memory (visual, spatial, and olfactory domains), and conditioning. | |
Reasoning and problem solving (cognitive symptoms) | Tests of behavioural flexibility in response to dynamic environments include set shifting and reversal learning. | |
Prepulse inhibition (sensorimotor deficits) | A reduction in the acoustic startle response is typically observed if the startling stimulus is preceded close in time by a low-intensity prepulse. | |
Anxiety-like behaviour | Patterns of exploration in environments such as the elevated plus maze, open field, or light-dark box. | |
Morphological/physiological | Enlarged lateral ventricles | Measurements of the volume of lateral ventricles using in vivo imaging techniques such as magnetic resonance imaging or postmortem analysis. |
Morphology of cortical, striatal, thalamic, and limbic areas | Includes measures of size/volume of brain areas, numbers of neurons, dendritic changes, and synaptic proteins. | |
Myelination | Assessment of the extent of myelination, particularly of cortical axons. | |
Glial cells | Changes in microglia, oligodendrocytes, and astrocytes are commonly studied. | |
Integrity of extracellular matrix | Alterations in structures such as perineuronal nets and proteins, including reelin. | |
Neurotransmitter-related alterations | Effects on dopamine, glutamate, serotonin (5-HT), GABA (notably parvalbumin-containing interneurons), glycine, D-serine, and neuroactive steroids. | |
Patterns of brain activity | Neural activity, dopamine neuron, activity, and patterns of oscillatory activity are measured using in vitro and in vivo electrophysiology techniques. |
While these measures relate to schizophrenia, it should be noted that none are exclusive for the disorder.16 See in-text citations for specific references to the measures listed.
GABA, gamma-aminobutyric acid; 5-HT, 5-hydroxytryptamine; NMDA, N-methyl-D-aspartate.
Developmental Models
Although the symptoms of schizophrenia typically appear in adolescence and early adulthood, there is now an extensive body of research linking schizophrenia to prenatal events such as obstetric complications or maternal infection during pregnancy.38,39 Thus, developmental animal models of schizophrenia involve environmental manipulations and/or drug administration during the perinatal and/or early postnatal period. The offspring are then studied over the course of development.5 One of the first developmental preparations used to study schizophrenia involved neonatal lesions of the rat ventral hippocampus on postnatal day 7 via local injection of either the excitotoxin ibotenic acid or temporary inactivation via tetrodotoxin. Neonatal lesions, but not similar lesions conducted in early adulthood, lead to an array of abnormalities consistent with schizophrenia that emerge at various times after puberty,40,41 similar to the typical onset of schizophrenia in late adolescence/early adulthood. Postnatal developmental manipulations involving administration of glutamate receptor agonists/antagonists, postweaning stress, or postweaning social isolation have also been used to model schizophrenia.33,42–44 Notably, postnatal social isolation results in behavioural changes in adulthood, including spontaneous locomotor hyperactivity, enhanced response to novelty, deficits in sensorimotor gating, cognitive impairment, and increased anxiety and aggression.5
The methylazomethanol (MAM) model involves prenatal administration of MAM into pregnant rats.45,46 MAM is an antimitotic and antiproliferative agent that methylates DNA47 and acts specifically on proliferation of neuroblasts without affecting glia or producing teratogenic effects in peripheral organs.48 Administering MAM to pregnant rats results in neuroanatomical, electrophysiological, and behavioural changes in the offspring that depend on the gestational day (GD) of administration, with GD17 animals being the most commonly used in schizophrenia animal models.49–51 Changes in MAM offspring include decreased volume of the medial prefrontal cortex, increased volume of lateral and third ventricles, decreases in parvalbumin interneuron markers in limbic and cortical areas, increased dopamine neuron activity in the ventral tegmental area, increased locomotor response to amphetamine and MK-801, decreased social interaction, decreased PPI, impaired memory, and increased anxiety (reviewed in Moore et al.45).
A second prenatal developmental strategy involves inducing maternal immune activation (MIA) in pregnant rodents with agents like the viral mimetic polyinosinic-polycytidylic acid (poly I: C) and studying the consequences for the offspring.52–62 A rather large body of evidence indicates that the offspring of rodents and primates that have undergone MIA during pregnancy show anatomical, neurochemical, electrophysiological, and behavioural changes consistent with schizophrenia,63–69 as well as other neurodevelopmental disorders, including autism.70 The maternal immune response appears critical for the effects of MIA on the offspring, with roles for maternal interleukin-671 and interleukin-17a72,73 identified. Recent work using the model has identified novel pathological changes in the offspring. For example, recent findings using this animal model implicate changes in parvalbumin interneurons and their associated perineuronal nets (extracellular matrix structures involved in structural and synaptic plasticity) in the prefrontal cortex (PFC) and hippocampal formation in cognitive deficits,69,74,75 consistent with studies on postmortem tissue from individuals with schizophrenia.74–76 Using mice, Shin Yim and colleagues77 showed that abnormalities in the primary somatosensory cortex underlie behavioural changes in the offspring following MIA.
Neurodevelopmental models permit investigations of the processes leading to the development of schizophrenia during the prodromal phase of the disease and potential development of prophylactic treatments to prevent the progression to psychosis.78 Combined, recent work in the MAM, MIA, neonatal ventral hippocampal lesion, and postweaning stress and isolation models has shown that interventions conducted around the time of adolescence alleviate some of the brain and behaviour changes in the animals.79–85 These interventions include cognitive training, antipsychotic administration, antioxidant administration, and benzodiazepine administration. Moreover, these models allow unique opportunities to create “2-hit” models,86 in which development is challenged during 2 critical periods (e.g., early brain development and then adolescence) in an effort to trigger a phenotype consistent with the development of schizophrenia later in life. While a single model is not sufficient to confirm efficacy for novel therapies, these well-validated neurodevelopmental models replicate many of the behavioural and neuroanatomical phenotypes of schizophrenia.87,88 Each of these components of schizophrenia can then be targeted for pharmacotherapy independently with separate molecules or specifically designed multitarget drugs,89 with the models providing accurate readouts of treatment efficacy.
Drug-Induced Models
The drugs used most frequently in animal models of schizophrenia are dopamine enhancers (e.g., amphetamine and apomorphine) and noncompetitive NMDA receptor antagonists (phencyclidine [PCP], ketamine, and MK-801). Amphetamine-induced psychosis in humans is well known and is proposed to result from the increased synaptic dopamine produced by this drug. Repeated, intermittent administration of amphetamine in rodents results in a persistent sensitization typified by exaggerated hyperactivity in response to an acute amphetamine challenge.90 However, repeated amphetamine does not induce social interaction deficits (which are proposed to be equivalent to negative symptoms in schizophrenia)91 and appears to affect only some cognitive tasks related to PFC function while leaving hippocampal function relatively unaffected.90 In summary, amphetamine induces psychotic-like changes but does not reliably produce negative or cognitive symptoms. In fact, some have proposed that treating selected patients with psychostimulants in an effort to improve negative symptoms is worth exploring.92
Hypofunctioning of NMDA glutamate receptors in schizophrenia has received considerable interest in recent years. Hence, noncompetitive NMDA receptor antagonists (ketamine, PCP, MK-801) have been administered to rodents in the hope that they would provide useful animal models of schizophrenia.93–98 In humans, PCP induces hallucinations and delusions, progressive withdrawal and poverty of speech, and impaired cognitive performance.99–101 In rodents, acute PCP produces hyperlocomotion (proposed to have translational relevance to positive symptoms in humans), social withdrawal, impaired PPI, and cognitive deficits.102–104 Effects of new antipsychotic candidates such as positive allosteric modulators of metabotropic glutamate receptors, the tetrahydroprotoberberine govadine, and nitric oxide donors (e.g., sodium nitroprusside [SNP]) have been tested using acute MK-801–induced disruptions of touchscreen-based tasks relevant to schizophrenia. These novel treatments reverse deficits on tasks, including paired associates learning and trial-unique, delayed nonmatching-to-location tasks.105–108
NMDA receptor activation results in the production of neuronal nitric oxide (NO). Bujas-Bobanovic and colleagues109 studied this relationship and found that PCP-induced effects in rats were blocked by the NO donor SNP. Hallak et al.110 translated these findings to patients and found that SNP, when administered by intravenous (IV) perfusion to patients with schizophrenia undergoing antipsychotic drug therapy, showed signs of improvement on scales measuring symptom severity within 4 hours. Notably, the improvements persisted for at least a week after a single dose of SNP. In healthy subjects, SNP was able to prevent psychogenic symptoms induced by ketamine.111 Other studies by the same group in rats showed that a single dose of SNP prevented psychosis-like behaviour produced by ketamine112 and that SNP treatment more consistently decreased ketamine-induced behaviours than other NO-related compounds.113 Thus, in these SNP studies, basic science research findings in animal models were translated to the clinical situation, then reverse translated back to animal models to further investigate mechanisms of action and other potential therapies. Such bidirectional translation is essential to developing new insights into the pathophysiology of schizophrenia and new avenues for treatment.
There has been a great deal of interest in the possible role of the amino acids glycine and D-serine, both coagonists at the NMDA receptor, in the aetiology and pharmacotherapy of schizophrenia. D-serine has a higher affinity than glycine for the serine glycine binding site on the GluN1 subunit of the NMDA receptor, and the focus of research has been more on D-serine in recent years. This emphasis will no doubt increase with recent withdrawal of glycine reuptake inhibitors from clinical trials.114 D-serine is formed from L-serine via serine racemase and catabolized by D-amino acid oxidase (DAO), and there is a great deal of interest in studies in rodents in which these enzymes are inhibited or induced by drugs or are knocked out, knocked down, or overexpressed in transgenic mice115,116 in an effort to identify new drugs that may be effective antipsychotics. Genetic studies on D-serine will be discussed in a later section of this review.
Other models have used local injections of drugs into specific brain areas in an effort to mimic a known pathophysiology associated with schizophrenia. For example, alterations of parvalbumin-containing GABAergic interneuron number and function in the prefrontal cortex is strongly associated with schizophrenia117 and a number of animal models of the disorder.74 Generally, these alterations are thought to result in reduced output of these neurons, thereby disturbing circuit function. In an effort to mimic this state in rodents, Tse and colleagues118 microinjected the GABA-A receptor antagonist bicuculline into rodent medial PFC and assessed cognition. Alterations in attentional processes, working memory, speed of processing, cognitive flexibility, emotion regulation, and locomotor activity consistent with a schizophrenia-like phenotype were observed.
Genetic Models
A genetic contribution to schizophrenia has long been a focus of study and the basis for several animal models. Numerous candidate genes have been associated with an increased risk of developing schizophrenia, and most of these genes relate to proteins relevant to neuronal plasticity, glutamatergic or dopaminergic function, and synaptogenesis.118–120 The largest genome-wide association study thus far, including data from 37,000 patients and 113,000 controls, was published in 2014.121 From this data set, 108 loci associated with schizophrenia were identified. Many of these genes have been used to generate animal models of schizophrenia, and new loci will allow for more models in mice. Notably, the identification of candidate genes is the first step in the generation of more sophisticated animal models of schizophrenia that incorporate the complex gene-by-environment interactions and gene-gene interactions that underlie the disease in humans.122 Several of the most established models based on genetic mutations are discussed below.
DISC1 (disrupted-in-schizophrenia 1) is a gene for DISC1, a synaptic protein expressed early in development and involved in pre- and postnatal development in neurons. DISC1 is considered a highly penetrant mutation and a risk factor for schizophrenia, although the genetic evidence for this claim has been debated.123 Mice with inducible and/or partial loss of DISC1 function have brain morphology consistent with schizophrenia, including enlarged ventricles and reduced cortical thickness and brain volume.124 Reduced immunoreactivity for parvalbumin, a marker of fast-spiking inhibitory interneurons, has also been observed in the medial PFC and hippocampus, consistent with postmortem studies in humans.124 While hyperactivity and impairments in volitional behaviour have been observed, there is some controversy about the degree to which these animals show changes in PPI, locomotor activity, or differences in negative symptoms and cognitive abilities.125–128
22q11.2 deletion syndrome is a genetic syndrome resulting from a deletion on chromosome 22q11.2.129 These deletions are associated with schizophrenia in humans and have been used to generate animal models of schizophrenia. It has been estimated that 22q11.2 deletions account for approximately 1% of cases of schizophrenia, with some studies suggesting as many as 22.6% of those with the deletions are diagnosed with schizophrenia or schizoaffective disorder.130 The core symptoms, treatment response, cognitive deficits, and brain pathologies do not appear to differ in 22q11.2 deletions relative to those diagnosed with schizophrenia that do not have the deletions. The mouse model recapitulates the major neuroanatomical findings observed in 22q11.2 deletion carriers,131 with alterations in cortical and subcortical grey matter volumes, enlarged ventricles, and reduced spine size and dendritic complexity.132 Behavioural studies in mice with 22q11.2 microdeletions reveal deficits in PPI, fear conditioning, impaired spatial memory, and deficits in delayed nonmatching to place tasks.133
Mutations in the genes for the cell adhesion molecule neuregulin 1 (NRG1) and its receptor ErbB4 also confer increased risk for schizophrenia.134–136 The NRG1 gene regulates expression of several isoforms of NRG1 that are involved in the development of the nervous system.137 Homozygous knockout (KO) in mice is lethal, but viable heterozygous, hypomorphic conditioned KOs can modulate neuregulin-ErbB4 signaling.138 These mice exhibit a behavioural phenotype with increased activity and exploration, and some reports noting working memory impairments in tests of spontaneous alternation138,139 in NRG1 hypomorphic mice, but not Erb4 hypomorphs, show reduced PPI. Notably, NRG1 hypomorphs have significantly fewer functional NMDA receptors than wild-type mice and reduced dendritic spine density on pyramidal neurons of the hippocampus.
Dysbindin is a synaptic protein implicated in the regulation of exocytosis, vesicle biogenesis, and receptor trafficking processes involved in excitatory synaptic transmission.140 A reduction in dysbindin expression affects NMDA receptor function and impairs working memory. Genetic variations in the gene encoding dysbindin impair cognition in humans and increase the risk for schizophrenia.140–145 Moreover, dysbindin gene and protein expression are reduced in the hippocampus and PFC of schizophrenic patients. Dysbindin mutant mice show behaviours consistent with a schizophrenia-like phenotype, including hyperactivity, learning and memory deficits, and increased impulsive and compulsive behaviours. Moreover, the mutants have disrupted dopamine/D2 signaling and altered neuronal excitability.146–149
Reelin is involved in synaptic formation and plasticity, and reduced reelin messenger RNA (mRNA) and its corresponding protein have been reported in cerebellum, hippocampus, and frontal cortex of individuals with schizophrenia.5 Deletion of 1 allele encoding the reelin gene produces a heterozygous mutant mouse.150 These mice have some schizophrenia-like pathology, including increased neuronal packing and reduced dendritic spine density in the frontal cortex and hippocampus.151 However, there is considerable controversy about the behavioural relevance of this model to schizophrenia, with variable findings in studies of social interaction, cognitive deficits, locomotor response, and PPI.5
Several genes showing up in association studies on schizophrenia are related to D-serine and include G72 (or DAP activator), DAO, and SRR (the gene for serum racemase).115,116 Insertion of the human G72 gene in mice produces a model that mimics some behavioural and cognitive characteristics of schizophrenia.152 The genes for the enzymes involved in the formation (serine racemase) and catabolism (DAO) of D-serine can be manipulated relatively easily. Behavioural effects noted in mice in which serine racemase is inhibited (leading to decreased brain levels of D-serine) indicate that this situation results in behavioural effects corresponding to symptoms in humans with schizophrenia.153 Conversely, genetic inactivation of DAO enhances extinction and reversal learning, suggestive of improvement of schizophrenia-like symptoms.154 It should also be mentioned that research now indicates that D-serine has antidepressant properties and may be a biomarker for nonresponse to the antidepressant effects of ketamine.155,156 Similarly, there is now extensive literature supporting GABAergic dysfunction in schizophrenia, including its interactions with dopamine and some neuroactive steroids.157 With regard to animal models, deficiency of glutamate decarboxylase 67, the enzyme responsible for the conversion of glutamate to GABA in mice, results in a schizophrenia-like phenotype.158
Sex Differences in Animal Models of Schizophrenia
There are clear sex differences with respect to age of onset, symptoms, and treatment/drug response in schizophrenia.159 Sex differences also exist between males and females in animal models of schizophrenia.160,161 These differences have been detected in both neurodevelopmental and genetic models of schizophrenia. Male- or female-specific deficits in PPI in offspring after prenatal PolyI: C have been reported, while deficits in both sexes were found in other studies.162,163 Similarly, male-specific deficits have been reported in studies of fear conditioning,164 set shifting,165 and recognition memory,166 while female-specific impairments have been reported in some, but not all, studies of spatial memory in polyI: C offspring.167 In general, prefrontal cortex–dependent tasks, including latent inhibition,168 PPI,169 recognition memory,170 and social behaviour,162 tend to be impaired to a greater extent in males than in females after MIA. These sex differences may parallel altered glutamate, aspartate and taurine concentrations, and parvalbumin-expressing inhibitory interneuron cell density in the prefrontal cortex of males but not females.168 Interestingly, MIA with polyI: C may preferentially disrupt prefrontal cortex–dependent tasks, and these deficits may only manifest in adulthood, potentially due to the delayed maturation of prefrontal cortex or puberty-related hormonal changes.160 However, statistical power (small group sizes per sex) and timing of polyI: C injection vary between studies and are important considerations in interpreting variable sex differences between polyI: C studies.160 Sex differences have also been reported in a neurodevelopmental model using prenatal exposure to the bacterial toxin lipopolysaccharide (LPS). Wischhof et al.57 reported that males, but not females, exhibited prolonged PPI deficits and impaired recognition memory. Interestingly, this study also reported deficient myelination in corticolimbic areas and reduced parvalbumin expression in the prefrontal cortex and hippocampus.
Overall, data on sex differences in genetic models of schizophrenia in the literature are variable and dependent on adequate sample size.160 For example, studies investigating mutations in DISC1 have found that when mutations are induced postnatally, sex-specific behavioural differences are observed, with males exhibiting hyperactivity and altered social interaction while females are impaired in spatial memory.170 Pre- and postnatal mutant expression results in increased aggression, altered social behaviour, and sensitivity to psychostimulant-induced locomotor hyperactivity in males and more depressive phenotypes in females.171 Male-specific increases in the ratio of excitatory to inhibitory synaptic activity in cortical pyramidal cells have also been reported in DISC1 mutants.172 Notably, mice with a naturally occurring mutation in DISCI show a similar phenotype to the inducible mutants, with male-specific hyperactivity. However, in these mutants, it was males that exhibited a depressive phenotype.173 This variability is common in studies of DISC1, and approximately half of studies to date have reported no sex differences in cognitive tasks, depressive behaviours, or locomotor activity.154
Variable sex differences have also been observed in other genetic models. Overall, cognitive impairments tend to be more severe in males in NRG1-ErbB4 mutants.160 Male-specific deficits in spatial learning in the Morris water maze have also been reported,174 but many studies lack statistical power to adequately detect sex differences.160 As such, further investigation of sex-specific effects on mutations affecting NRG1-ErbB4 is required. While most studies including both sexes of reelin mutants lacked sufficiently large samples to differentiate between sexes, differences have been observed in studies of heterozygous reelin mutant mice with larger samples.175 Adult male, but not female, heterozygous reelin mutant mice have enhanced locomotor response to MK-801175 as well as deficits in social recognition memory. In virtually all cases, additional studies with adequate sample size for both sexes are required to better define sex differences in genetic models.
Glial Contributions to Schizophrenia
Glial cell dysfunction has been implicated in several animal models of schizophrenia.176–178 Activated microglia are an important part of the immune system and, by releasing proinflammatory cytokines, may contribute to symptoms seen in animal models based on immune reactions.54–56,179 Genetic mutation of the genes encoding CX3C chemokine receptor 1 (CX3CR1) fractalkine (CX3CL1) and its receptor (CX3CR1), which are concentrated in microglia and involved in immune cell trafficking to the central nervous system, induces a schizophrenia-like phenotype.180,181 Similarly, genetic and proteomic studies in humans have suggested that abnormalities in the cytokines interleukin-6, interleukin-1B, interferon-Y, tumour necrosis factor α, and the major histocompatibility complex are associated with schizophrenia.182–186 Genetic and transcriptomic studies of S100B and glial fibrillary acid protein, both highly expressed in astrocytes, reveal altered expression in the plasma and brains of individuals with schizophrenia.187,188 There is now also substantial evidence, using demyelination animal models such as rodents treated with cuprizone, that oligodendrocyte function and demyelination may also contribute to the symptoms of schizophrenia.189,190 Oligodendrocyte-related genes are frequently identified as associated with schizophrenia in genetic, transcriptomic, and proteomic studies.191 Consistent with this, a genomic analysis of 10 publicly available gene sets identified a glial-oligodendrocyte pathway as significantly associated with schizophrenia, suggesting that these myelination abnormalities in schizophrenia are at least in part due to inherited factors.192 Thus, modeling of schizophrenia involves the integration of genetic factors across multiple cell types, neurodevelopmental processes, and complex gene-by-environment interactions.
Summary
Despite the complicated, heterogeneous aetiology of schizophrenia, which almost certainly includes contributions from both environmental and genetic factors, animal models have increased our knowledge of the brain pathology in schizophrenia. Moreover, these models allow for identification of pathways for pharmacotherapy and improved screening and validation of potential novel antipsychotics. With continued refinement of the models and our understanding of the pathophysiology of schizophrenia, the development of more rapidly acting therapies with reduced side effect profiles compared to the agents currently available is possible.
Acknowledgements
The authors are grateful for funding from the University of Alberta, the University of São Paulo, and the University of Saskatchewan. John G. Howland’s research in this area is supported by operating grants from the Canadian Institutes for Health Research (125984 and 153111). Ian R. Winship’s research in this area is supported by operating grants from the Canadian Institutes for Health Research (153111), the Natural Science and Engineering Research Council (RGPIN-2017-05380), the University Hospital Foundation, and Alberta Innovates Health Solutions.
Footnotes
Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Funding: The author(s) received no financial support for the research, authorship, and/or publication of this article.
ORCID iD: Joao Paulo Maia-de-Oliveira, MD, PhD http://orcid.org/0000-0002-7637-2498
References
- 1. Saha S, Chant D, Welham J, et al. A systematic review of the prevalence of schizophrenia. PLoS Med. 2005;2(5):e141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Yang AC, Tsai SJ. New targets for schizophrenia treatment beyond the dopamine hypothesis. Int J Mol Sci. 2017;18(8). pii: E1689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Salavati B, Rajji TK, Price R, et al. Imaging-based neurochemistry in schizophrenia: a systematic review and implications for dysfunctional long-term potentiation. Schizophr Bull. 2015;41(1):44–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Balu DT, Coyle JT. The NMDA receptor ‘glycine modulatory site’ in schizophrenia: D-serine, glycine, and beyond. Curr Opin Pharmacol. 2015;20:109–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Jones CA, Watson DJ, Fone KC. Animal models of schizophrenia. Br J Pharmacol. 2011;164(4):1162–1194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Canetta S, Kellendonk C. Can we use mice to study schizophrenia? Philos Trans R Soc Lond B Biol Sci. 2018;373(1742). pii: 20170032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Young JW, Powell SB, Risbrough V, et al. Using the MATRICS to guide development of a preclinical cognitive test battery for research in schizophrenia. Pharmacol Ther. 2009;122(2):150–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Hvoslef-Eide M, Mar AC, Nilsson SR, et al. The NEWMEDS rodent touchscreen test battery for cognition relevant to schizophrenia. Psychopharmacology. 2015;232(21-22):3853–3872. [DOI] [PubMed] [Google Scholar]
- 9. Bussey TJ, Holmes A, Lyon L, et al. New translational assays for preclinical modelling of cognition in schizophrenia: the touchscreen testing method for mice and rats. Neuropharmacology. 2012;62(3):1191–1203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Nithianantharajah J, McKenchanie AG, Stewart TJ, et al. Bridging the translational divide: identical cognitive touchscreen testing in mice and humans carrying mutations in a disease-relevant homologous gene. Sci Reports. 2015;5:14613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Cuthburt BN, Insel TR. Toward the future of psychiatric diagnosis: the seven pillars of RdoC. BMC Med. 2013;11:126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Kozak MJ, Cuthbert BN. The NIMH research domain criteria initiative: background, issues, and pragmatics. Psychophysiology. 2016;53(3):286–297. [DOI] [PubMed] [Google Scholar]
- 13. Morris SE, Vaidyanathan U, Cuthbert BN. Changing the diagnostic concept of schizophrenia: the NIMH research domain criteria initiative. Nebraska Symposium Motivation. 2016;63:225–252. [DOI] [PubMed] [Google Scholar]
- 14. Kaffman A, Krystal JH. New frontiers in animal research of psychiatric illness. Methods Mol Biol. 2012;829:3–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Young JW, Winstanley CA, Brady AM, et al. Research Domain Criteria versus DSM V: how does this debate affect attempts to model corticostriatal dysfunction in animals? Neurosci Biobehav Rev. 2017;76(Pt B):301–316. [DOI] [PubMed] [Google Scholar]
- 16. Nestler EJ, Hyman SE. Animal models of neuropsychiatric disorders. Nat Neurosci. 2010;13(10):1161–1169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Macedo DS, Araujo DP, Sampaio LRL, et al. Animal models of prenatal immune challenge and their contribution to the study of schizophrenia: a systematic review. Braz J Med Biol Res. 2012;45(3):179–186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Barnes SA, Der-Avakian A, Markou A. Anhedonia, avolition, and anticipatory deficits: assessments in animals with relevance to the negative symptoms of schizophrenia. Eur Neuropsychopharmacol. 2014;24(5):744–758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Bickel S, Javitt DC. Neurophysiological and neurochemical animal models of schizophrenia: focus on glutamate. Behav Brain Res. 2009;204(2):352–362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Feifel D, Shilling PD. Promise and pitfalls of animal models of schizophrenia. Curr Psychiatry Rep. 2010;12(4):327–334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Flores G, Morales-Medina JC, Diaz A. Neuronal and brain morphological changes in animal models of schizophrenia. Behav Brain Res. 2016;301:190–203. [DOI] [PubMed] [Google Scholar]
- 22. Geyer MA, Olivier B, Joels M, et al. From antipsychotic to anti-schizophrenia drugs: role of animal models. Trends Pharmacol Sci. 2012;33(10):515–521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Green AR, Gabrielsson J, Fone KCF. Translational neuropharmacology and the appropriate and effective use of animal models. Br J Pharmacol. 2011;164(4):1041–1043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Kannan G, Sawa A, Pletnikov MV. Mouse models of gene-environment interactions in schizophrenia. Neurobiol Dis. 2013;57:5–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Greek R, Menache A. Systematic reviews of animal models: methodology versus epistemology. Int J Med Sci. 2013;10(3):206–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Lin CY, Sawa A, Jaaro-Peled H. Better understanding of mechanisms of schizophrenia and bipolar disorder: from human gene expression profiles to mouse models. Neurobiol Dis. 2012;45(1):48–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Mattei D, Schweibold R, Wolf SA. Brain in flames—animal models of psychosis: utility and limitations. Neuropsychiatr Dis Treat. 2015;11:1313–1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Miyamoto Y, Nitta A. Behavioral phenotypes for negative symptoms in animal models of schizophrenia. J Pharmacol Sci. 2014;126(4):310–320. [DOI] [PubMed] [Google Scholar]
- 29. Burrows EL, Hannan AJ. Cognitive endophenotypes, gene-environment interactions and experience-dependent plasticity in animal models of schizophrenia. Biol Psychol. 2016;116:82–89. [DOI] [PubMed] [Google Scholar]
- 30. 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] [PubMed] [Google Scholar]
- 31. Powell CM, Miyakawa T. Schizophrenia-relevant behavioral testing in rodent models: a uniquely human disorder? Biol Psychiatry. 2006;59(12):1198–1207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Powell SB. Models of neurodevelopmental abnormalities in schizophrenia. Curr Top Behav Neurosci. 2010;4:435–481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Puhl MD, Mintzopoulos D, Jensen JE, et al. In vivo magnetic resonance studies reveal neuroanatomical and neurochemical abnormalities in the serine racemase knockout mouse model of schizophrenia. Neurobiol Dis. 2015;73:269–274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Severance EG, Prandovszky E, Castiglione J, et al. Gastroenterology issues in schizophrenia: why the gut matters. Curr Psychiatry Rep. 2015;17(5):27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Wischhof L, Irrsack E, Osorio C, et al. Prenatal LPS-exposure—a neurodevelopmental rat model of schizophrenia—differentially affects cognitive functions, myelination and parvalbumin expression in male and female offspring. Prog NeuroPsychopharmacol Biol Psychiatry. 2015;57:17–30. [DOI] [PubMed] [Google Scholar]
- 36. Wong AH, Josselyn SA. Caution when diagnosing your mouse with schizophrenia: the use and misuse of model animals for understanding psychiatric disorders. Biol Psychiatry. 2016;79(1):32–38. [DOI] [PubMed] [Google Scholar]
- 37. Yee BK, Singer P. A conceptual and practical guide to the behavioural evaluation of animal models of the symptomatology and therapy of schizophrenia. Cell Tissue Res. 2013;354(1):221–246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Brown AS, Derkits EJ. Prenatal infection and schizophrenia: a review of epidemiologic and translational studies. Am J Psychiatry. 2010;167(3):261–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Brown AS. Epidemiologic studies of exposure to prenatal infection and risk of schizophrenia and autism. Dev Neurobiol. 2012;72(10):1272–1276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Becker A, Grecksch G, Bernstein HG, et al. Social behavior in rats lesioned with ibotenic acid in the hippocampus: quantitative and qualitative analysis. Psychopharmacology. 1999;144(4):333–338. [DOI] [PubMed] [Google Scholar]
- 41. Tseng KY, Chambers RA, Lipska BK. The neonatal ventral hippocampal lesion as a heuristic neurodevelopmental model of schizophrenia. Behav Brain Res. 2009;204(2):295–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Harris LW, Sharp T, Gartlon J, et al. Long-term behavioural, molecular and morphological effects of neonatal NMDA receptor antagonism. Eur J Neuorsci. 2003;18(6):1706–1710. [DOI] [PubMed] [Google Scholar]
- 43. Kjaerby C, Hovelso N, Dalby NO, et al. Phencyclidine administration during neurodevelopment alters network activity in prefrontal cortex and hippocampus in adult rats. J Neurophysiol. 2017;118(2):1002–1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Howland JG, Hannesson DK, Phillips AG. Delayed onset of prepulse inhibition deficits following kainic acid treatment on postnatal day 7 in rats. Eur J Neurosci. 2004;20(10):2639–2648. [DOI] [PubMed] [Google Scholar]
- 45. Moore H, Jentsch JD, Ghajarnia M, et al. A neurobehavioral systems analysis of adult rats exposed to methylazoxymethanol acetate on E17: implications for the neuropathology of schizophrenia. Biol Psychiatry. 2006;60(3):253–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Lodge DJ, Grace AA. Gestational methylazoxymethanol acetate administration: a developmental disruption model of schizophrenia. Behav Brain Res. 2009;204(2):306–312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Matsumot H, Higa HH. Studies on methylazoxymethanol aglycone of cycasin: methylation of nucleic acids in vitro. Biochem J. 1966;98(2):20C–22C. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Cattabeni F, DiLuca M. Developmental models of brain dysfunctions induced by targeted cellular ablations with methylazoxymethanol. Physiol Rev. 1997;77(1):199–215. [DOI] [PubMed] [Google Scholar]
- 49. Talamini LM, Ellenbroek B, Koch T, et al. Impaired sensory gating and attention in rats with developmental abnormalities of the mesocortex—implications for schizophrenia. Ann N Y Acad Sci. 2000;911:486–494. [DOI] [PubMed] [Google Scholar]
- 50. Gomes FV, Rincón-Cortés M, Grace AA. Adolescence as a period of vulnerability and intervention in schizophrenia: insights from the MAM model. Neurosci Biobehav Rev. 2016;70:260–270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Modinos G, Allen P, Grace AA, et al. Translating the MAM model of psychosis to humans. Trends Neurosci. 2015;38(3):129–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Feigenson KA, Kusnecov AW, Silverstein SM. Inflammation and the two-hit hypothesis of schizophrenia. Neurosci Biobehav Rev. 2014;38:72–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Ibi D, Yamada K. Therapeutic targets for neurodevelopmental disorders emerging from animal models with perinatal immune activation. Int J Mol Sci. 2015;16(12):28218–28229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Macedo DS, Araujo DP, Sampaio LR, et al. Animal models of prenatal immune challenge and their contribution to the study of schizophrenia: a systematic review. Braz J Med Biol Res. 2012;45(3):179–186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Muller N, Weidinger E, Leitner B, et al. The role of inflammation in schizophrenia. Front Neurosci. 2015;9:372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Reisinger S, Khan D, Kong E, et al. The poly(I: C)–induced maternal immune activation model in preclinical neuropsychiatric drug discovery. Pharmacol Ther. 2015;149:213–226. [DOI] [PubMed] [Google Scholar]
- 57. Wischhof L, Irrsack E, Osorio C, et al. Prenatal LPS-exposure—a neurodevelopmental rat model of schizophrenia—differentially affects cognitive functions, myelination and parvalbumin expression in male and female offspring. Prog Neuropsychopharmacol Biol Psychiatry. 2015;57:17–30. [DOI] [PubMed] [Google Scholar]
- 58. Fineberg AM, Ellman LM. Inflammatory cytokines and neurological and neurocognitive alterations in the course of schizophrenia. Biol Psychiatry. 2013;73(10):951–966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Jenkins T. Perinatal complications and schizophrenia: involvement of the immune system. Front Neurosci. 2013;110:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Feigenson KA, Kusnecov AW, Silverstein SM. Inflammation and the two-hit hypothesis of schizophrenia. Neurosci Biobehav Rev. 2014;38:72–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Möller M, Swanepoel T, Harvey BH. Neurodevelopmental animal models reveal the convergent role of neurotransmitter systems, inflammation, and oxidative stress as biomarkers of schizophrenia: implications for novel drug development. ACS Chem Neurosci. 2015;6(7):987–1016. [DOI] [PubMed] [Google Scholar]
- 62. Müller N, Weidinger E, Leitner B, et al. The role of inflammation in schizophrenia. Front Neurosci. 2015;9:372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Meyer U. Prenatal poly (I: C) exposure and other developmental immune activation models in rodent systems. Biol Psychiatry. 2014;75:307–315. [DOI] [PubMed] [Google Scholar]
- 64. Dickerson DD, Overeem KA, Wolff AR, et al. Association of aberrant neural synchrony and altered GAD67 expression following exposure to maternal immune activation, a risk factor for schizophrenia. Transl Psychiatry. 2014;4(7):e418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Howland JG, Cazakoff BN, Zhang Y. Altered object-in-place recognition memory, prepulse inhibition, and locomotor activity in the offspring of rats exposed to a viral mimetic during pregnancy. Neurosci. 2012;201:184–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Piontkewitz Y, Arad M, Weiner I. Tracing the development of psychosis and its prevention: what can be learned from animal models. Neuropharmacology. 2012;62(3):1273–1289. [DOI] [PubMed] [Google Scholar]
- 67. Zhang Y, Cazakoff BN, Thai CA, et al. Prenatal exposure to a viral mimetic alters behavioural flexibility in male, but not female, rats. Neuropharmacology. 2012;62(3):1299–1307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Machado CJ, Whitaker AM, Smith SE, et al. Maternal immune activation in nonhuman primates alters social attention in juvenile offspring. Biol Psychiatry 2015;77(9):823–832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Paylor JW, Lins BR, Greba Q, et al. Developmental disruption of perineuronal nets in the medial prefrontal cortex after maternal immune activation. Sci Rep. 2016,6:37580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Careaga M, Murai T, Bauman MD. Maternal immune activation and autism spectrum disorder: from rodents to nonhuman and human primates. Biol Psychiatry. 2017;81(5):391–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Smith SE, Li J, Garbett K, et al. Maternal immune activation alters fetal brain development through interleukin-6. J Neurosci. 2007;27(40):10695–10702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Choi GB, Yim YS, Wong H, et al. The maternal interleukin-17a pathway in mice promotes autism-like phenotypes in offspring. Science. 2016;351(6276):933–939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Kim S, Kim H, Yim YS, et al. Maternal gut bacteria promote neurodevelopmental abnormalities in mouse offspring. Nature. 2017;549(7673):528–532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Steullet P, Cabungcal JH, Coyle J, et al. Oxidative stress-driven parvalbumin interneuron impairment as a common mechanism in models of schizophrenia. Mol Psychiatry. 2017;22(7):936–943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Giovanoli S, Weber L, Meyer U. Single and combined effects of prenatal immune activation and peripubertal stress on parvalbumin and reelin expression in the hippocampal formation. Brain Behav Immunity. 2014;40:48–54. [DOI] [PubMed] [Google Scholar]
- 76. Careaqa M, Murai T, Bauman MD. Maternal immune activation and autism spectrum disorder: from rodents to nonhuman and human primates. Biol Psychiatry. 2017;81(5):391–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Shin Yim Y, Park A, Berrios J, et al. Reversing behavioural abnormalities in mice exposed to maternal inflammation. Nature. 2017;549(7673):482–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Powell SB, Risbrough VB, Geyer MA. Potential use of animal models to examine anti-psychotic prophylaxis for schizophrenia. Clin Neurosci Res. 2003;3:289–296. [Google Scholar]
- 79. Pantazopoulos H, Markota M, Jaquet F, et al. Aggrecan and chondroitin-6-sulfate abnormalities in schizophrenia and bipolar disorder: a postmortem study on the amygdala. Transl Psychiatry 2015;5(1):e496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Tseng KY, Chambers RA, Lipsla BK. The neonatal ventral hippocampal lesion as a heuristic neurodevelopmental model of schizophrenia. Behav Brain Res. 2009;204(2):295–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Chambers RA, Moore J, McEvoy JP, et al. Cognitive effects of neonatal hippocampal lesions in a rat model of schizophrenia. Neuropsychopharmacology. 1996;15(6):587–594. [DOI] [PubMed] [Google Scholar]
- 82. Gomes FV, Rincon-Cortes M, Grace AA. Adolescence as a period of vulnerability and intervention in schizophrenia: insights from the MAM model. Neurosci Biobehav Rev. 2016;70:260–270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Piontkewitz Y, Arad M, Weiner I. Tracing the development of psychosis and its prevention: what can be learned from animal models. Neuropharmacology. 2012;62(3):1273–1289. [DOI] [PubMed] [Google Scholar]
- 84. Patterson PH. Immune involvement in schizophrenia and autism: etiology, pathology and animal models. Behav Brain Res. 2009;204(2):313–331. [DOI] [PubMed] [Google Scholar]
- 85. Fone KCF, Porkess MV. Behavioural and neurochemical effects of post-weaning social isolation in rodents—relevance to developmental psychiatric disorders. Neurosci Biobehav Rev. 2008;32(6):1087–1101. [DOI] [PubMed] [Google Scholar]
- 86. Keshavan MS, Hogarty GE. Brain maturational processes and delayed onset in schizophrenia. Psychopathology. 1999;11(3):525–543. [DOI] [PubMed] [Google Scholar]
- 87. Wilson C, Terry AV., Jr Neurodevelopmental animal models of schizophrenia: role in novel drug discovery and development. Clin Schizophr Relat Psychoses. 2010;4(2):124–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Powell SB. Models of neurodevelopmental abnormalities in schizophrenia. Curr Top Behav Neurosci. 2010;4:435–481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Gründer G, Hippius H, Carlsson A. The ‘atypicality’ of antipsychotics: a concept re-examined and re-defined. Nat Rev Drug Discov. 2009;8(3):197–202. [DOI] [PubMed] [Google Scholar]
- 90. Featherstone RE, Rizos Z, Kapur S, et al. A sensitizing regimen of amphetamine that disrupts attentional set-shifting does not disrupt working or long-term memory. Behav Brain Res. 2008;189(1):170–179. [DOI] [PubMed] [Google Scholar]
- 91. Sams-Dodd F. A test of the predictive validity of animal models of schizophrenia based on phencyclidine and d-amphetamine. Neuropsychopharmacology. 1998;18(4):293–304. [DOI] [PubMed] [Google Scholar]
- 92. Lindenmayer JP, Nasrallah H, Pucci M, et al. A systematic review of psychostimulant treatment of negative symptoms of schizophrenia: challenges and therapeutic opportunities. Schizoph Res. 2013;147(2-3):241–252. [DOI] [PubMed] [Google Scholar]
- 93. Grayson B, Barnes SA, Markou A, et al. Postnatal phencyclidine (PCP) as a neurodevelopmental animal model of schizophrenia pathophysiology and symptomatology: a review. Curr Top Behav Neurosci. 2016;29:403–428. [DOI] [PubMed] [Google Scholar]
- 94. Janhunen SK, Svard H, Talpos J, et al. The subchronic phencyclidine rat model: relevance for the assessment of novel therapeutics for cognitive impairment associated with schizophrenia. Psychopharmacol (Berl). 2015;232(21-22):4059–4083. [DOI] [PubMed] [Google Scholar]
- 95. Jeevakumar V, Driskill C, Paine A, et al. Ketamine administration during the second postnatal week induces enduring schizophrenia-like behavioral symptoms and reduces parvalbumin expression in the medial prefrontal cortex of adult mice. Behav Brain Res. 2015;282:165–175. [DOI] [PubMed] [Google Scholar]
- 96. Jeevakumar V, Driskill C, Paine A, et al. Ketamine administration during the second postnatal week induces enduring schizophrenia-like behavioral symptoms and reduces parvalbumin expression in the medial prefrontal cortex of adult mice. Behav Brain Res. 2015;282:165–175. [DOI] [PubMed] [Google Scholar]
- 97. Lodge D, Mercier MS. Ketamine and phencyclidine: the good, the bad and the unexpected. Br J Pharmacol. 2015;172(17):4254–4276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Kocsis B, Brown RE, McCarley RW, et al. Impact of ketamine on neuronal network dynamics, translational modeling of schizophrenia-relevant deficits. CNS Neurosci Ther. 2013;19(6):437–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Luby ED, Cohen BD, Rosenbaum G, et al. Study of a new schizophrenomimetic drug, sernyl. AMA Arch Neurol Psychiatry. 1959;81(3):363–369. [DOI] [PubMed] [Google Scholar]
- 100. Steinpreis RE. The behavioral and neurochemical effects of phencyclidine in humans and animals: some implications for modeling psychoses. Behav Brain Res. 1996;74(1-2):45–55. [DOI] [PubMed] [Google Scholar]
- 101. Javitt DC, Zukin SR. Recent advances in the phencyclidine model of schizophrenia. Am J Psychiatry. 1991;148(10):1301–1308. [DOI] [PubMed] [Google Scholar]
- 102. Kalinichev M, Robbins MJ, Hartfield EM, et al. Comparison between intraperitoneal and subcutaneous phencyclidine administration in Sprague-Dawley rats: a locomotor activity and gene induction study. Prog Neuropsychopharmacol Biol Psychiatry. 2007;32(2):414–422. [DOI] [PubMed] [Google Scholar]
- 103. Sams-Dodd F. Distinct effects of d-amphetamine and phencyclidine on the social behaviour of rats. Behav Pharmacol. 1995;6(1):55–65. [PubMed] [Google Scholar]
- 104. Mansbach RS, Geyer MA. Effects of phencyclidine and phencyclidine biologs on sensorimotor gating in the rat. Neuropsychopharmacology. 1989;2(4):299–308. [DOI] [PubMed] [Google Scholar]
- 105. Lins BR, Howland JG. Effects of the metabotropic glutamate receptor 5 positive allosteric modulator CDPPB on rats tested with the paired associates learning task in touchscreen-equipped operant conditioning chambers. Behav Brain Res. 2016;301:152–160. [DOI] [PubMed] [Google Scholar]
- 106. Lins BR, Phillips AB, Howland JG. Effects of D- and L-govadine on the disruption of touchscreen object-location paired associates learning in rats by acute MK-801 treatment. Psychopharmacology. 2015;232(23):4371–4382. [DOI] [PubMed] [Google Scholar]
- 107. Lins BR, Marks WN, Phillips AG, et al. Dissociable effects of the d- and l- enantiomers of govadine on the disruption of prepulse inhibition by MK-801 and apomorphine in male Long-Evans rats. Psychopharmacology. 2017;234(7):1079–1091. [DOI] [PubMed] [Google Scholar]
- 108. Hurtubise JL, Marks WN, Davis DA, et al. MK-801–induced impairments on the trial-unique, delayed nonmatching-to-location task in rats: effects of acute sodium nitroprusside. Psychopharmacology. 2017;234(2):211–222. [DOI] [PubMed] [Google Scholar]
- 109. Bujas-Bobanovic M, Bird DC, Robertson HA, et al. Blockade of phencyclidine-induced effects by a nitric oxide donor. Br J Pharmacol. 2000;130(5):1005–1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Hallak JE, Maia-de-Oliveira JP, Abrao J, et al. Rapid improvement of acute schizophrenia symptoms after intravenous sodium nitroprusside: a randomized double-blind, placebo-controlled trial. JAMA Psychiatry. 2013;70(7):668–676. [DOI] [PubMed] [Google Scholar]
- 111. Rezende TMN, Maia-de-Oliveira JP, Kandratavicius L, et al. Effects of sodium nitroprusside in the prevention of schizophrenia-like symptoms induced by ketamine—a translational double-blind study. Arch Clin Psychiatry. 2017;44(6):149–153. [Google Scholar]
- 112. Maia-de-Oliveira JP, Lobao-Soares B, Ramalho T, et al. Nitroprusside single-dose prevents the psychosis-like behavior induced by ketamine in rats for up to one week. Schizophr Res. 2015;162(1-3):211–215. [DOI] [PubMed] [Google Scholar]
- 113. Kandratavicius L, Balista PA, Wolf DC, et al. Effects of nitric oxide–related compounds in the acute ketamine animal model of schizophrenia. BMC Neurosci. 2015;16:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Thomas R, Baker G, Dursun S, et al. Glycine reuptake inhibitors in the treatment of negative symptoms of schizophrenia. Bull Clin Psychopharmacol. 2014;24(3):195–200. [Google Scholar]
- 115. Nunes E, MacKenzie EM, Rossolatos D, et al. D-serine and schizophrenia: an update. Expert Rev Neurotherapeutics. 2012;12(7):801–812. [DOI] [PubMed] [Google Scholar]
- 116. Labrie V, Wong AH, Roder JC. Contributions of the D-serine pathway to schizophrenia. Neuropharmacology. 2012;62(3):1484–1503. [DOI] [PubMed] [Google Scholar]
- 117. Gonzalez-Burgos G, Cho RY, Lewis DA. Alterations in cortical network oscillations and parvalbumin neurons in schizophrenia. Biol Psychiatry. 2015;77(12):1031–1040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Tse MT, Piantadosi PT, Floresco SB. Prefrontal cortical gamma-aminobutyric acid transmission and cognitive function: drawing links to schizophrenia from preclinical research. Biol Psychiatry. 2015;77(11):929–939. [DOI] [PubMed] [Google Scholar]
- 119. Harrison PJ, Weinberger DR. Schizophrenia genes, gene expression, and neuropathology: on the matter of their convergence. Mol Psychiatry. 2002;10(1):40–68. [DOI] [PubMed] [Google Scholar]
- 120. Farrell MS, Werge T, Sklar P, et al. Evaluating historical candidate genes for schizophrenia. Mol Psychiatry. 2015;20(5):555–562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Schizophrenia Working Group of the Psychiatric Genomics Consortium 2014.6
- 122. McOmish CE, Burrows EL, Hannan AJ. Identifying novel interventional strategies for psychiatric disorders: integrating genomics, ‘enviromics’ and gene-environment interactions in valid preclinical models. Br J Pharmacol. 2014;171(20):4719–4728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Sullivan PF. Questions about DISC1 as a genetic risk factor for schizophrenia. Mol Psychiatry. 2013;18(10):1050–1052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Jaaro-Peled H. Gene models of schizophrenia: DISC1 mouse models. Prog Brain Res. 2009;179:75–86. [DOI] [PubMed] [Google Scholar]
- 125. Clapcote S, Lipina T, Millar J, et al. Behavioral phenotypes of Disc1 missense mutations in mice. Neuron. 2007;54(3):387–402. [DOI] [PubMed] [Google Scholar]
- 126. Hikida T, Jaaro-Peled H, Seshadr IS, et al. Dominant-negative DISC1 transgenic mice display schizophrenia-associated phenotypes detected by measures translatable to humans. Proc Natl Acad Sci USA. 2007;104(36):14501–14506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Kvajo M, McKellar H, Arguello P, et al. A mutation in mouse Disc1 that models a schizophrenia risk allele leads to specific alterations in neuronal architecture and cognition. Proc Natl Acad Sci USA. 2008;105:7076–7081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Lipina TV, Zai C, Hlousek D, et al. Maternal immune activation during gestation interacts with Disc1 point mutation to exacerbate schizophrenia-related behaviours in mice. J Neurosci. 2013;33(18):7654–7666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Bassett AS, Chow EWC. Schizophrenia and 22q11.2 deletion syndrome. Curr Psychiatry Reports. 2008;10(2):148–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Bassett AS, Chow EW, Husted J, et al. Clinical features of 78 adults with 22q11 deletion syndrome. Am J Med Gen. 2005;138(4):307–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Ellegood J, Markx S, Lerch JP, et al. Neuroanatomical phenotypes in a mouse model of the 22q11.2 microdeletion. Mol Psychiatry. 2014;19(1):99–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Stark KL, Xu B, Bagchi A, et al. Altered brain microRNA biogenesis contributes to phenotypic deficits in a 22q11-deletion mouse model. Nat Genet. 2008;40(6):751–760. [DOI] [PubMed] [Google Scholar]
- 133. Karayiorgou M, Simon TJ, Gogos JA. 22q11.2 Microdeletions: linking DNA structural variation to brain dysfunction and schizophrenia. Nat Rev Neurosci. 2010;11(6):402–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. O’Tuathaigh CM, O’Sullivan GJ, Kinsella A, et al. Sexually dimorphic changes in the exploratory and habituation profiles of heterozygoud neuregulin-1 knockout mice. Neuroreport. 2006;17(1):79–83. [DOI] [PubMed] [Google Scholar]
- 135. Stefansson H, Sigurdsson E, Steinthorsdottir V, et al. Neuregulin 1 and susceptibility to schizophrenia. Am J Hum Genet. 2002;71(4):877–892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Karl T, Duffy L, Scimone A, et al. Altered motor activity, exploration and anxiety in heterozygous neuregulin 1 mutant mice: implications for understanding schizophrenia. Genes Brain Behav. 2007;6(7):677–687. [DOI] [PubMed] [Google Scholar]
- 137. Engel M, Snikeris P, Jenner A, et al. Neuregulin 1 prevents phencyclidine-induced behavioral impairments and disruptions to GABAergic signaling in mice. Int J Neuropsychopharmacol. 2015;18(7):pyu114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Mei L, Xiong W. Neuregulin 1 in neural development, synaptic plasticity and schizophrenia. Nat Rev Neurosci. 2008;9(6):437–452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Chen Y-J, Johnson MA, Lieberman MD, et al. Type III neuregulin-1 is required for normal sensorimotor gating, memory-related behaviors, and corticostriatal circuit components. J Neurosci. 2008;28(27):6872–6883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Burdick KE, Lencz T, Funke B, et al. Genetic variation in DTNBP1 influences general cognitive ability. Hum Mol Genet. 2006;15(10):1563–1568. [DOI] [PubMed] [Google Scholar]
- 141. Fallgatter AJ, Herrmann MJ, Hohoff C, et al. DTNBP1 (dysbindin) gene variants modulate prefrontal brain function in healthy individuals. Neuropsychopharmacology. 2006;31(9):2002–2010. [DOI] [PubMed] [Google Scholar]
- 142. Talbot K, Eidem WL, Tinsley CL, et al. Dysbindin-1 is reduced in intrinsic, glutamatergic terminals of the hippocampal formation in schizophrenia. J Clin Invest. 2004;113(9):1353–1363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Weickert CS, Rothmond DA, Hyde TM, et al. Reduced DTNBP1 (dysbindin-1) mRNA in the hippocampal formation of schizophrenia patients. Schizo Res. 2008;98(1-3):105–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Weickert CS, Straub RE, McClintock BW, et al. Human dysbindin (DTNBP1) gene expression in normal brain and in schizophrenic prefrontal cortex and midbrain. Arch Gen Psychiatry. 2004;61(6):544–55. [DOI] [PubMed] [Google Scholar]
- 145. Tang J, LeGros RP, Louneva N, et al. Dysbindin-1 in dorsolateral prefrontal cortex of schizophrenia cases is recued in an isoform-specific manner unrelated to dysbindin-1 mRNA expression. Hum Mol Genet. 2009;18(20):3851–3863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Cox MM, Tucker AM, Tang J, et al. Neurobehavioral abnormalities in the dysbindin-1 mutant, sandy, on a C57BL/6 genetic background. Genes Brain Behav. 2009;8(4):390–397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Papaleo F, Yang F, Garcia S, et al. Dysbindin-1 modulates prefrontal cortical activity and schizophrenia-like behaviours via dopamine/D2 pathways. Mol Psychiatry. 2012;17(1):85–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Carr GV, Jenkins KA, Weinberger DR, et al. Loss of dysbindin-1 in mice impairs reward-based operant learning by increasing impulsive and compulsive behavior. Behav Brain Res. 2013;241:173–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Karlsgodt K, Robleto K, Trantham-Davidson H, et al. Reduced dysbindin expression mediates N-methyl-D-aspartate receptor hypofunction and impaired working memory performance. Biol Psychiatry. 2011;69(1):28–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Liu W, Pesold C, Rodriguez M, et al. Down-regulation of dendritic spine and glutamic acid decarboxylase 67 expressions in the reelin haploinsufficient heterozygous reeler mouse. Proc Natl Acad Sci USA. 2001;98(6):3477–3482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Kruger D, Howell J, Hebert B, et al. Assessment of cognitive function in the heterozygous reeler mouse. Psychopharmacology (Berl). 2006;189(1):95–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Otte DM, Bilkei-Gorzó A, Filiou MD, et al. Behavioral changes in G72/G30 transgenic mice. Eur Neuropsychopharmacol. 2009;19(5):339–348. [DOI] [PubMed] [Google Scholar]
- 153. Labrie V, Fukumura R, Rastogi A, et al. Serine racemase is associated with schizophrenia susceptibility in humans and in a mouse model. Hum Mol Genet. 2009;18(17):3227–3243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Labrie V, Duffy S, Wang W, et al. Genetic inactivation of D-amino acid oxidase enhances extinction and reversal learning in mice. Learn Mem. 2008;16(1):28–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Malkesman O, Austin DR, Tragon T, et al. Acute D-serine treatment produces antidepressant-like effects in rodents. Neuropsychopharmacology. 2012;15(8):1135–1148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Hashimoto K. Blood D-serine levels as a predictive biomarker for the rapid antidepressant effects of the NMDA receptor antagonist ketamine. Psychopharmacology. 2014;231(20):4081–4082. [DOI] [PubMed] [Google Scholar]
- 157. Wassef A, Baker J, Kochan LD. GABA and schizophrenia: a review of basic science and clinical studies. J Clin Psychopharmacol. 2003;23(6):601–640. [DOI] [PubMed] [Google Scholar]
- 158. Fujihara K, Miwa H, Kakizaki T, et al. Glutamate decarboxylase 67 deficiency in a subset of GABAergic neurons induces schizophrenia-related phenotypes. Neuropsychopharmacology. 2015;40(10):2475–2486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Goldstein JM, Cherkerzian S, Tsuang MT, et al. Sex differences in the genetic risk for schizophrenia: history of the evidence for sex-specific and sex-dependent effects. Am J Med Genet B Neuropsychiatr Genet. 2013;162B(7):698–710. [DOI] [PubMed] [Google Scholar]
- 160. Hill RA. Sex differences in animal models of schizophrenia shed light on the underlying pathopsychology. Neurosci Behav Rev. 2016;67:41–56. [DOI] [PubMed] [Google Scholar]
- 161. Kekesi G, Petrovszki Z, Benedek G, et al. Sex-specific alterations in behavioral and cognitive functions in a “three hit” animal model of schizophrenia. Behav Brain Res. 2015;284:85–93. [DOI] [PubMed] [Google Scholar]
- 162. O’Leary C, Desbonnet L, Clarke N, et al. 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] [PubMed] [Google Scholar]
- 163. Ratnayake U, Quinn T, LaRosa DA, et al. Prenatal exposure to the viral mimetic poly I: C alters fetal brain cytokine expression and postnatal behavior. Dev Neurosci. 2014;36(2):83–94. [DOI] [PubMed] [Google Scholar]
- 164. Vorhees CV, Graham DL, Braun AA, et al. Prenatal immune challenge in rats: altered responses to dopaminergic and glutamatergic agents, prepulse inhibition of acoustic startle, and reduced route-based learning as a function of maternal body weight gain after prenatal exposure to poly IC. Synapse. 2012;66(8):725–737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Zhang Y, Cazakoff BN, Thai CA, et al. Prenatal exposure to a viral mimetic alters behavioural flexibility in male, but not female, rats. Neuropharmacology. 2012;62(3):1299–1307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Howland JG, Cazakoff BN, Zhang Y. Altered object-in-place recognition memory, prepulse inhibition, and locomotor activity in the offspring of rats exposed to a viral mimetic during pregnancy. Neuroscience. 2012;201:184–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. O’Leary C, Desbonnet L, Clarke N, et al. 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] [PubMed] [Google Scholar]
- 168. Nelson AJ, Thur KE, Marsden CA, et al. Catecholaminergic depletion within the prelimbic medial prefrontal cortex enhances latent inhibition. Neuroscience. 2010;170(1):99–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Hill RA. Sex differences in animal models of schizophrenia shed light on the underlying pathophysiology. Neurosci Biobehav Rev. 2016;67:41–56. [DOI] [PubMed] [Google Scholar]
- 170. Pletnikov MV, Ayhan Y, Nikolskaia O, et al. Inducible expression of mutant human DISC1 in mice is associated with brain and behavioral abnormalities reminiscent of schizophrenia. Mol Psychiatry. 2008;13(2):173–186. [DOI] [PubMed] [Google Scholar]
- 171. Ayhan Y, Abazyan B, Nomura J, et al. Differential effects of prenatal and postnatal expressions of mutant human DISC1 on neurobehavioral phenotypes in transgenic mice: evidence for neurodevelopmental origin of major psychiatric disorders. Mol Psychiatry. 2011;16(3):293–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Holley SM, Wang EA, Cepeda C, et al. Frontal cortical synaptic communication is abnormal in Disc1 genetic mouse models of schizophrenia. Schizophr Res. 2013;146(1-3):264–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Gómez-Sintes R, Kvajo M, Gogos JA, et al. Mice with a naturally occurring DISC1 mutation display a broad spectrum of behaviors associated to psychiatric disorders. Front Behav Neurosci. 2014;8:253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Golub MS, Germann SL, Lloyd KC. Behavioral characteristics of a nervous system- specific erbB4 knock-out mouse. Behav Brain Res. 2004;153(1):159–170. [DOI] [PubMed] [Google Scholar]
- 175. van den Buuse M, Halley P, Hill R, et al. Altered N-methyl-D-aspartate receptor function in reelin heterozygous mice: male-female differences and comparison with dopaminergic activity. Prog Neuropsychopharmacol Biol Psychiatry. 2012;37(2):237–246. [DOI] [PubMed] [Google Scholar]
- 176. Xia M, Abazyan S, Jouroukhin Y, et al. Behavioral sequelae of astrocyte dysfunction: focus on animal models of schizophrenia. Schizophrenia Res. 2016;176(1):72–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Wang C, Aleksic B, Ozaki N. Glia-related genes and their contribution to schizophrenia. Psychiatry Clin Neurosci. 2015;69(8):448–461. [DOI] [PubMed] [Google Scholar]
- 178. Bilimoria PM, Stevens B. Microglia function during brain development: new insights from animal models. Brain Res. 2015;1617:7–17. [DOI] [PubMed] [Google Scholar]
- 179. Aricioglu F, Ozkartal CS, Unal G, et al. Neuroinflammation in schizophrenia: a critical review and the future. Bull Clin Psychopharmacol. 2016;26(4):429–437. [Google Scholar]
- 180. Rogers JT, Morganti JM, Bachstetter AD, et al. CX#CR1 deficiency leads to impairment of hippocampal cognitive function and synaptic plasticity. J Neurosci. 2011;31(45):16241–16250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Reshef R, Kreisel T, Beroukhim KD, et al. Microglia and their CX3CR1 signaling are involved in hippocampal- but not olfactory bulb–related memory and neurogenesis. Brain Behav Immun. 2014;41:239–250. [DOI] [PubMed] [Google Scholar]
- 182. Fillman SG, Cloonan N, Catts VS, et al. Increased inflammatory markers identified in the dorsolateral prefrontal cortex of individuals with schizophrenia. Mol Psychiatry. 2013;18(2):206–214. [DOI] [PubMed] [Google Scholar]
- 183. Mondelli V, Cattaneo A, Murri MB, et al. Stress and inflammation reduce brain-derived neurotrophic factor expression in first-episode psychosis: a pathway to smaller hippocampal volume. J Clin Psychiatry. 2011;72(12):1677–1684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Meisenzahl EM, Rujescu D, Kirner A, et al. Association of an interleukin-1beta genetic polymorphism with altered brain structure in patients with schizophrenia. Am J Psychiatry. 2001;158(8):1316–1319. [DOI] [PubMed] [Google Scholar]
- 185. Paul-Samojedny M, Owczarek A, Suchanek R, et al. Association study of interferon gamma (IFN-y) + 874T/A gene polymorphism in patients with paranoid schizophrenia. J Mol Neurosci. 2011;43(3):309–315. [DOI] [PubMed] [Google Scholar]
- 186. Miller BJ, Buckley P, Seabolt W, et al. Meta-analysis of cytokine alterations in schizophrenia: clinical status and antipsychotic effects. Biol Psychiatry. 2011;70(7):663–671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Katsel P, Byne W, Roussos P, et al. Astrocyte and glutamate markers in the superficial, deep, and white matter layers of the anterior cingulate gyrus in schizophrenia. Neuropsychopharmacology. 2011;36(6):1171–1177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Sugai T, Kawamura M, Iritani S, et al. Prefrontal abnormality of schizophrenia revealed by DNA microarray: impact on glial and neurotrophic gene expression. Ann N Y Acad Sci. 2004;1025:84–91. [DOI] [PubMed] [Google Scholar]
- 189. Chew LJ, Fusar-Poli P, Schmitz T. Oligodendroglial alterations and the role of microglia in white matter injury: relevance to schizophrenia. Dev Neurosci. 2013;35(2-3):102–129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Xu H, Li XM. White matter abnormalities and animal models examining a putative role of altered white matter in schizophrenia. Schizophr Res Treat. 2011;2011:826976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Wang C, Aleksic B, Ozaki N. Glia-related genes and their contribution to schizophrenia. Psychiatry Clin Neurosci. 2015;69(8):448–461. [DOI] [PubMed] [Google Scholar]
- 192. Duncan LE, Holmans PA, Lee PH, et al. Pathway analyses implicate glial cells in schizophrenia. PLoS One. 2014;9(2):e89441. [DOI] [PMC free article] [PubMed] [Google Scholar]