Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
PLoS One ; 18(12): e0295855, 2023.
Article in English | MEDLINE | ID: mdl-38127959

ABSTRACT

Early life stress (ELS) can impact brain development and is a risk factor for neurodevelopmental disorders such as schizophrenia. Post-weaning social isolation (SI) is used to model ELS in animals, using isolation stress to disrupt a normal developmental trajectory. We aimed to investigate how SI affects the expression of genes in mouse hippocampus and to investigate how these changes related to the genetic basis of neurodevelopmental phenotypes. BL/6J mice were exposed to post-weaning SI (PD21-25) or treated as group-housed controls (n = 7-8 per group). RNA sequencing was performed on tissue samples from the hippocampus of adult male and female mice. Four hundred and 1,215 differentially-expressed genes (DEGs) at a false discovery rate of < 0.05 were detected between SI and control samples for males and females respectively. DEGS for both males and females were significantly overrepresented in gene ontologies related to synaptic structure and function, especially the post-synapse. DEGs were enriched for common variant (SNP) heritability in humans that contributes to risk of neuropsychiatric disorders (schizophrenia, bipolar disorder) and to cognitive function. DEGs were also enriched for genes harbouring rare de novo variants that contribute to autism spectrum disorder and other developmental disorders. Finally, cell type analysis revealed populations of hippocampal astrocytes that were enriched for DEGs, indicating effects in these cell types as well as neurons. Overall, these data suggest a convergence between genes dysregulated by the SI stressor in the mouse and genes associated with neurodevelopmental disorders and cognitive phenotypes in humans.


Subject(s)
Autism Spectrum Disorder , Adult , Humans , Male , Animals , Mice , Female , Gene Expression Profiling , Hippocampus/metabolism , Social Isolation , Synapses , Phenotype , Risk Factors , Human Genetics
2.
Behav Brain Res ; 430: 113930, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35609792

ABSTRACT

Evidence suggests that early life adversity, such as maternal immune activation (MIA), can alter brain development in the offspring and confer increased risk for psychopathology and psychiatric illness in later life. In this study, the long-term effects of MIA, post-weaning social isolation, and the combination were assessed on behavioural and immunological profiles in adult male and female offspring. On gestation day 12.5, pregnant mice were weighed and injected with either polyinosinic:polycytidylic acid (5 mg/kg) or saline and cytokines levels were assayed 3 hrs later to confirm immune activation. The behaviour and immunological profiles of male and female offspring were examined in adolescence (P34-36), and adulthood (P55-80). MIA induced an increase in the pro-inflammatory cytokine IL-6 in pregnant dams three hours after administration (p < 0.001) that correlated with a decrease in body temperature (p < 0.05). The effect of MIA on the immunological phenotype of the offspring was evident in adolescence, but not in adulthood. MIA selectively induced hypoactivity in adolescent males, a phenotype that persisted until adulthood, but had no effect on cognition in males or females. In contrast, social isolation stress from adolescence resulted in impaired sociability (p < 0.05) and increased anxiety (p < 0.05) particularly in adult females. There was no synergistic effect of the dual-hit on immune parameters, sociability, anxiety or cognitive behaviours. Given the negative impact and sex-dependent effects of SI stress on locomotor and anxiety-like behaviour, future investigations should examine whether the health risks of social isolation, such as that experience during the COVID-19 pandemic, are mediated through increased anxiety.


Subject(s)
COVID-19 , Prenatal Exposure Delayed Effects , Schizophrenia , Adolescent , Adult , Animals , Behavior, Animal/physiology , Cytokines/pharmacology , Disease Models, Animal , Endophenotypes , Female , Humans , Male , Mice , Pandemics , Poly I-C/pharmacology , Pregnancy , Social Isolation , Weaning
3.
Genes (Basel) ; 12(9)2021 08 30.
Article in English | MEDLINE | ID: mdl-34573345

ABSTRACT

Maternal immune activation (MIA) is a known risk factor for schizophrenia (SCZ) and autism spectrum disorder (ASD) and is often modelled in animal studies in order to study the effect of prenatal infection on brain function including behaviour and gene expression. Although the effect of MIA on gene expression are highly heterogeneous, combining data from multiple gene expression studies in a robust method may shed light on the true underlying biological effects caused by MIA and this could inform studies of SCZ and ASD. This study combined four RNA-seq and microarray datasets in an overlap analysis and ranked meta-analysis in order to investigate genes, pathways and cell types dysregulated in the MIA mouse models. Genes linked to SCZ and ASD and crucial in neurodevelopmental processes including neural tube folding, regulation of cellular stress and neuronal/glial cell differentiation were among the most consistently dysregulated in these ranked analyses. Gene ontologies including K+ ion channel function, neuron and glial cell differentiation, synaptic structure, axonal outgrowth, cilia function and lipid metabolism were also strongly implicated. Single-cell analysis identified excitatory and inhibitory cell types in the cortex, hippocampus and striatum that may be affected by MIA and are also enriched for genes associated with SCZ, ASD and cognitive phenotypes. This points to the cellular location of molecular mechanisms that may be consistent between the MIA model and neurodevelopmental disease, improving our understanding of its utility to study prenatal infection as an environmental stressor.


Subject(s)
Brain/physiology , Gene Expression , Neurodevelopmental Disorders/etiology , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/pathology , Animals , Brain/drug effects , Disease Models, Animal , Female , Gene Expression/drug effects , Gene Ontology , Humans , Mice , Neurodevelopmental Disorders/pathology , Poly I-C/pharmacology , Pregnancy , Prenatal Exposure Delayed Effects/genetics
4.
Neurosci Lett ; 736: 135218, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32615248

ABSTRACT

Dysbindin-1 is implicated in several aspects of schizophrenia, including cognition and both glutamatergic and dopaminergic neurotransmission. Targeted knockout of dysbindin-1A (Dys-1A KO), the most abundant and widely expressed isoform in the brain, is associated with deficits in delay/interference-dependent working memory. Using an ethologically based approach, the following behavioural phenotypes were examined in Dys-1A KO mice: exploratory activity, social interaction, anxiety and problem-solving ability. Levels of monoamines and their metabolites were measured in striatum, hippocampus and prefrontal cortex using high-performance liquid chromatography with electrochemical detection. The ethogram of initial exploration in Dys-1A KO mice was characterised by increased rearing from a seated position; over subsequent habituation, stillness was decreased relative to wildtype. In a test of dyadic social interaction with an unfamiliar conspecific in a novel environment, female KO mice showed an increase in investigative social behaviours. Marble burying behaviour was unchanged. Using the puzzle-box test to measure general problem-solving performance, no effect of genotype was observed across nine trials of increasing complexity. Dys-1A KO demonstrated lower levels of 5-HT in ratio to its metabolite 5-HIAA in the prefrontal cortex. These studies elaborate the behavioural and neurochemical phenotype of Dys-1A KO mice, revealing subtle genotype-related differences in non-social and social exploratory behaviours and habituation of exploration in a novel environment, as well as changes in 5-HT activity in brain areas related to schizophrenia.


Subject(s)
Behavior, Animal/physiology , Brain/metabolism , Dysbindin/metabolism , Schizophrenia/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Isoforms , Serotonin/metabolism
5.
J Psychopharmacol ; 33(12): 1610-1619, 2019 12.
Article in English | MEDLINE | ID: mdl-31556815

ABSTRACT

BACKGROUND: As exposure to stress has been linked to the onset and maintenance of psychotic illness, its pathogenesis may involve environmental stressors interacting with genetic vulnerability. AIM: To establish whether acute stress interacts with a targeted mutation of the gene encoding the neurodevelopmental factor dystrobrevin-binding protein 1 (DTNBP1), resulting in a specific loss of the isoform dysbindin-1A, to influence schizophrenia-relevant phenotypes in mice during adolescence and adulthood. METHODS: Male and female mice with a heterozygous or homozygous deletion of DTNBP1 were assessed in the open field test following acute restraint stress in adolescence (Day 35) and young adulthood (Day 60-70). Effects of acute restraint stress on memory retention in the novel object recognition test was also assessed in adulthood. Baseline corticosterone was measured in serum samples and, brain-derived neurotrophic factor (BDNF), glucocorticoid and mineralocorticoid receptor gene expression levels were measured in the hippocampus of adult mice. RESULTS: In the open field, deletion of dysbindin-1A induced hyperactivity and attenuated the action of stress to reduce hyperactivity in adolescence but not in adulthood; in females deletion of dysbindin-1A attenuated the effect of acute stress to increase anxiety-related behaviour in adolescence but not in adulthood. In the novel object recognition test, deletion of dysbindin-1A impaired memory and also revealed an increase in anxiety-related behaviour and a decrease in hippocampal BDNF gene expression in males. CONCLUSIONS: These data suggest that deletion of dysbindin-1A influences behaviours related to schizophrenia and anxiety more robustly in adolescence than in adulthood and that dysbindin-1A influences stress-related responses in a sex-dependent manner.


Subject(s)
Anxiety/psychology , Dysbindin/genetics , Schizophrenia/physiopathology , Stress, Psychological/psychology , Age Factors , Animals , Behavior, Animal/physiology , Brain-Derived Neurotrophic Factor/genetics , Cognition/physiology , Female , Gene Expression Regulation , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Schizophrenia/genetics , Sequence Deletion , Stress, Psychological/genetics
6.
Behav Brain Res ; 334: 50-54, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28736331

ABSTRACT

Increasing evidence points to a functional role of the enteric microbiota in brain development, function and behaviour including the regulation of transcriptional activity in the hippocampus. Changes in CNS miRNA expression may reflect the colonisation status of the gut. Given the pivotal impact of miRNAs on gene expression, our study was based on the hypothesis that gene expression would also be altered in the germ-free state in the hippocampus. We measured miRNAs in the hippocampus of Germ free (GF), conventional (C) and Germ free colonised (exGF) Swiss Webster mice. miRNAs were selected for follow up based on significant differences in expression between groups according to sex and colonisation status. The expression of miR-294-5p was increased in male germ free animals and was normalised following colonisation. Targets of the differentially expressed miRNAs were over-represented in the kynurenine pathway. We show that the microbiota modulates the expression of miRNAs associated with kynurenine pathway metabolism and, demonstrate that the gut microbiota regulates the expression of kynurenine pathway genes in the hippocampus. We also show a sex-specific role for the microbiota in the regulation of miR-294-5p expression in the hippocampus. The gut microbiota plays an important role in modulating small RNAs that influence hippocampal gene expression, a process critical to hippocampal development.


Subject(s)
Gastrointestinal Microbiome/physiology , Hippocampus/metabolism , Kynurenine/metabolism , MicroRNAs/metabolism , Animals , Female , Gene Expression Regulation , Male , Mice , MicroRNAs/genetics , Microbiota , Sex Characteristics
7.
Behav Brain Res ; 320: 113-118, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27916686

ABSTRACT

The complex genetic origins of many human disorders suggest that epistatic (gene×gene) interactions may contribute to a significant proportion of their heritability estimates and phenotypic heterogeneity. Simultaneous disruption of the developmental genes and schizophrenia risk factors Neuregulin-1 (NRG1) and Disrupted-in-schizophrenia 1 (DISC1) in mice has been shown to produce disease-relevant and domain-specific phenotypic profiles different from that observed following disruption of either gene alone. In the current study, anxiety and stress responsivity phenotypes in male and female mutant mice with simultaneous disruption of DISC1 and NRG1 were examined. NRG1×DISC1 mutant mice were generated and adult mice from each genotype were assessed for pain sensitivity (hot plate and tail flick tests), anxiety (light-dark box), and stress-induced hypothermia. Serum samples were assayed to measure circulating levels of pro-inflammatory cytokines. Mice with the NRG1 mutation, irrespective of DISC1 mutation, spent significantly more time in the light chamber, displayed increased core body temperature following acute stress, and decreased pain sensitivity. Basal serum levels of cytokines IL8, IL1ß and IL10 were decreased in NRG1 mutants. Mutation of DISC1, in the absence of epistatic interaction with NRG1, was associated with increased serum levels of IL1ß. Epistatic effects were evident for IL6, IL12 and TNFα. NRG1 mutation alters stress and pain responsivity, anxiety, and is associated with changes in basal cytokine levels. Epistasis resulting from synergistic NRG1 and DISC1 gene mutations altered pro-inflammatory cytokine levels relative to the effects of each of these genes individually, highlighting the importance of epistatic mechanisms in immune-related pathology.


Subject(s)
Cytokines/blood , Nerve Tissue Proteins/metabolism , Neuregulin-1/metabolism , Pain Threshold/physiology , Adaptation, Ocular , Analysis of Variance , Animals , Disease Models, Animal , Female , Fever/etiology , Hyperalgesia/metabolism , Male , Mice , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/genetics , Neuregulin-1/genetics , Pain Measurement , Sex Factors , Stress, Psychological/complications
8.
Schizophr Bull ; 43(1): 214-225, 2017 01.
Article in English | MEDLINE | ID: mdl-27613806

ABSTRACT

Few studies have addressed likely gene × gene (ie, epistatic) interactions in mediating risk for schizophrenia. Using a preclinical genetic approach, we investigated whether simultaneous disruption of the risk factors Neuregulin-1 (NRG1) and Disrupted-in-schizophrenia 1 (DISC1) would produce a disease-relevant phenotypic profile different from that observed following disruption to either gene alone. NRG1 heterozygotes exhibited hyperactivity and disruption to prepulse inhibition, both reversed by antipsychotic treatment, and accompanied by reduced striatal dopamine D2 receptor protein expression, impaired social cognition, and altered glutamatergic synaptic protein expression in selected brain areas. Single gene DISC1 mutants demonstrated a disruption in social cognition and nest-building, altered brain 5-hydroxytryptamine levels and hippocampal ErbB4 expression, and decreased cortical expression of the schizophrenia-associated microRNA miR-29b. Co-disruption of DISC1 and NRG1, indicative of epistasis, evoked an impairment in sociability and enhanced self-grooming, accompanied by changes in hypothalamic oxytocin/vasopressin gene expression. The findings indicate specific behavioral correlates and underlying cellular pathways downstream of main effects of DNA variation in the schizophrenia-associated genes NRG1 and DISC1.


Subject(s)
Behavior, Animal , Brain/metabolism , Endophenotypes , Epistasis, Genetic , Nerve Tissue Proteins/metabolism , Neuregulin-1/metabolism , Psychotic Disorders , Schizophrenia , Amphetamines/pharmacology , Animals , Disease Models, Animal , Female , Grooming , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Nesting Behavior , Neuregulin-1/genetics , Oxytocin/metabolism , Prepulse Inhibition/genetics , Psychomotor Agitation/genetics , Psychotic Disorders/genetics , Psychotic Disorders/metabolism , Psychotic Disorders/physiopathology , Schizophrenia/genetics , Schizophrenia/metabolism , Schizophrenia/physiopathology , Social Behavior , Vasopressins/metabolism
9.
Neural Plast ; 2016: 2173748, 2016.
Article in English | MEDLINE | ID: mdl-27725886

ABSTRACT

The study of gene × environment, as well as epistatic interactions in schizophrenia, has provided important insight into the complex etiopathologic basis of schizophrenia. It has also increased our understanding of the role of susceptibility genes in the disorder and is an important consideration as we seek to translate genetic advances into novel antipsychotic treatment targets. This review summarises data arising from research involving the modelling of gene × environment interactions in schizophrenia using preclinical genetic models. Evidence for synergistic effects on the expression of schizophrenia-relevant endophenotypes will be discussed. It is proposed that valid and multifactorial preclinical models are important tools for identifying critical areas, as well as underlying mechanisms, of convergence of genetic and environmental risk factors, and their interaction in schizophrenia.


Subject(s)
Gene-Environment Interaction , Schizophrenia/genetics , Animals , Disease Models, Animal , Genetic Predisposition to Disease , Humans , Risk Factors
11.
Psychoneuroendocrinology ; 60: 58-74, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26135201

ABSTRACT

Early-life adverse experiences, including prenatal stress (PNS), are associated with a higher prevalence of neurodevelopmental, cardiovascular and metabolic disorders in affected offspring. Here, in a rat model of chronic PNS, we investigate the impact of late gestational stress on physiological outcomes in adulthood. Sprague-Dawley pregnant dams were subjected to repeated restraint stress from embryonic day 14 to day 20, and their male offspring were assessed at 4 months of age. PNS induced an exaggeration of the hypothalamic-pituitary-adrenal (HPA) axis response to stress, as well as an elevation of blood pressure and impairment of cognitive function. Altered respiratory control was also observed, as demonstrated by increased variability in basal respiratory frequency and abnormal frequency responses to both hypoxic and hypercapnic challenges. PNS also affected gastrointestinal neurodevelopment and function, as measured by a decrease in the innervation density of distal colon and an increase in the colonic secretory response to catecholaminergic stimulation. Finally, PNS induced long lasting alterations in the intestinal microbiota composition. 16S rRNA gene 454 pyrosequencing revealed a strong trend towards decreased numbers of bacteria in the Lactobacillus genus, accompanied by elevated abundance of the Oscillibacter, Anaerotruncus and Peptococcus genera in PNS animals. Strikingly, relative abundance of distinct bacteria genera significantly correlated with certain respiratory parameters and the responsiveness of the HPA axis to stress. Together, these findings provide novel evidence that PNS induces long-term maladaptive alterations in the gastrointestinal and respiratory systems, accompanied by hyper-responsiveness to stress and alterations in the gut microbiota.


Subject(s)
Colon/microbiology , Colon/physiopathology , Gastrointestinal Microbiome , Prenatal Exposure Delayed Effects/physiopathology , Stress, Psychological/microbiology , Stress, Psychological/physiopathology , Animals , Blood Pressure , Cognition , Female , Hypothalamo-Hypophyseal System/physiopathology , Male , Motor Activity , Pituitary-Adrenal System/physiopathology , Pregnancy , Prenatal Exposure Delayed Effects/psychology , RNA, Bacterial/analysis , RNA, Ribosomal, 16S/metabolism , Rats , Rats, Sprague-Dawley , Respiratory Mechanics , Restraint, Physical , Social Behavior
12.
Brain Behav Immun ; 48: 165-73, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25866195

ABSTRACT

BACKGROUND: There is growing appreciation for the importance of bacteria in shaping brain development and behaviour. Adolescence and early adulthood are crucial developmental periods during which exposure to harmful environmental factors can have a permanent impact on brain function. Such environmental factors include perturbations of the gut bacteria that may affect gut-brain communication, altering the trajectory of brain development, and increasing vulnerability to psychiatric disorders. Here we assess the effects of gut bacterial depletion from weaning onwards on adult cognitive, social and emotional behaviours and markers of gut-brain axis dysfunction in mice. METHODS: Mice were treated with a combination of antibiotics from weaning onwards and effects on behaviours and potential gut-brain axis neuromodulators (tryptophan, monoamines, and neuropeptides) and BDNF expression were assessed in adulthood. RESULTS: Antibiotic-treatment depleted and restructured gut microbiota composition of caecal contents and decreased spleen weights in adulthood. Depletion of the gut microbiota from weaning onwards reduced anxiety, induced cognitive deficits, altered dynamics of the tryptophan metabolic pathway, and significantly reduced BDNF, oxytocin and vasopressin expression in the adult brain. CONCLUSIONS: Microbiota depletion from weaning onwards by means of chronic treatment with antibiotics in mice impacts on anxiety and cognitive behaviours as well as key neuromodulators of gut-brain communication in a manner that is similar to that reported in germ-free mice. This model may represent a more amenable alternative for germ-free mice in the assessment of microbiota modulation of behaviour. Finally, these data suggest that despite the presence of a normal gut microbiome in early postnatal life, reduced abundance and diversity of the gut microbiota from weaning influences adult behaviours and key neuromodulators of the microbiota-gut-brain axis suggesting that dysregulation of this axis in the post-weaning period may contribute to the pathogenesis of disorders associated with altered anxiety and cognition.


Subject(s)
Behavior, Animal/physiology , Brain/physiology , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/microbiology , Animals , Anti-Bacterial Agents/pharmacology , Behavior, Animal/drug effects , Body Weight/drug effects , Body Weight/physiology , Brain/drug effects , Brain/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Gastrointestinal Microbiome/drug effects , Mice , Tryptophan/metabolism
13.
Eur Neuropsychopharmacol ; 24(5): 800-21, 2014 May.
Article in English | MEDLINE | ID: mdl-24290531

ABSTRACT

Modelling negative symptoms in any animal model, particularly in mice mutant for genes related to schizophrenia, is complicated by the absence of the following key elements that might assist in developing validation criteria: clinical clarity surrounding this symptom constellation; any clear association between negative symptoms and pathological signature(s) in the brain; and therapeutic strategies with material clinical efficacy against these symptoms. In this review, the application of mutant mouse models to the study of negative symptoms is subjected to critical evaluation, focussing on the following challenges: (a) conceptual issues relating to negative symptoms and their evaluation in mutant models; (b) measurement of negative symptoms in mice, in terms of social behaviour, motivational deficits/avolition and anhedonia; (c) studies in mutants with disruption of genes either regulating aspects of neurotransmission implicated in schizophrenia or associated with risk for psychotic illness; (d) the disaggregation of behavioural phenotypes into underlying pathobiological processes, as a key to the development of new therapeutic strategies for negative symptoms. Advances in genetic and molecular technologies are facilitating these processes, such that more accurate models of putative schizophrenia-linked genetic abnormalities are becoming feasible. This progress in terms of mimicking the genetic contribution to distinct domains of psychopathology associated with psychotic illness must be matched by advances in conceptual/clinical relevance and sensitivity/specificity of phenotypic assessments at the level of behaviour.


Subject(s)
Disease Models, Animal , Mice, Transgenic , Phenotype , Psychotic Disorders/physiopathology , Schizophrenia/physiopathology , Schizophrenic Psychology , Animals , Humans , Psychotic Disorders/genetics , Psychotic Disorders/psychology , Schizophrenia/genetics
14.
Mamm Genome ; 25(1-2): 49-74, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24281320

ABSTRACT

Bacterial colonisation of the gut plays a major role in postnatal development and maturation of key systems that have the capacity to influence central nervous system (CNS) programming and signaling, including the immune and endocrine systems. Individually, these systems have been implicated in the neuropathology of many CNS disorders and collectively they form an important bidirectional pathway of communication between the microbiota and the brain in health and disease. Regulation of the microbiome-brain-gut axis is essential for maintaining homeostasis, including that of the CNS. Moreover, there is now expanding evidence for the view that commensal organisms within the gut play a role in early programming and later responsivity of the stress system. Research has focused on how the microbiota communicates with the CNS and thereby influences brain function. The routes of this communication are not fully elucidated but include neural, humoral, immune and metabolic pathways. This view is underpinned by studies in germ-free animals and in animals exposed to pathogenic bacterial infections, probiotic agents or antibiotics which indicate a role for the gut microbiota in the regulation of mood, cognition, pain and obesity. Thus, the concept of a microbiome-brain-gut axis is emerging which suggests that modulation of the gut microflora may be a tractable strategy for developing novel therapeutics for complex stress-related CNS disorders where there is a huge unmet medical need.


Subject(s)
Brain/physiology , Gastrointestinal Tract/microbiology , Microbiota , Animals , Behavior , Humans , Mental Disorders/etiology , Signal Transduction , Stress, Physiological , Stress, Psychological
15.
Expert Rev Clin Pharmacol ; 5(6): 667-76, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23234324

ABSTRACT

The vague relationship between diagnosis, underlying etiology and a rudimentary understanding of the pathophysiology of psychosis, particularly schizophrenia, has made it difficult to develop and validate suitable disease models for such disorders. Despite recent technological advancements, animal models have yet to yield a revolutionary treatment for schizophrenia. Refinement and standardization of assessment methods in the preclinical domain and streamlining of concepts from which animal models are generated are required, particularly in relation to models that recapitulate cognitive and negative symptoms of schizophrenia. In this review, caveats of current treatments for schizophrenia and current animal modeling strategies are examined in the context of their validity and potential for discovery of novel therapies, and finally, future prospects for the field are considered.


Subject(s)
Antipsychotic Agents/therapeutic use , Schizophrenia/drug therapy , Animals , Disease Models, Animal , Humans , Models, Neurological , Neuropsychological Tests , Schizophrenia/etiology , Schizophrenia/physiopathology , Schizophrenic Psychology
16.
Handb Exp Pharmacol ; (213): 113-45, 2012.
Article in English | MEDLINE | ID: mdl-23027414

ABSTRACT

In this review we consider the application of mutant mouse phenotypes to the study of psychotic illness in general and schizophrenia in particular, as they relate to behavioral, psychopharmacological, and cellular phenotypes of putative import for antipsychotic drug development. Mutant models appear to be heuristic at two main levels; firstly, by indicating the functional roles of neuronal components thought to be of relevance to the putative pathobiology of psychotic illness, they help resolve overt behavioral and underlying cellular processes regulated by those neuronal components; secondly, by indicating the functional roles of genes associated with risk for psychotic illness, they help resolve overt behavioral and underlying cellular processes regulated by those risk genes. We focus initially on models of dopaminergic and glutamatergic dysfunction. Then, we consider advances in the genetics of schizophrenia and mutant models relating to replicable risk genes. Lastly, we extend this discussion by exemplifying two new variant approaches in mutant mice that may serve as prototypes for advancing antipsychotic drug development. There is continuing need not only to address numerous technical challenges but also to develop more "real-world" paradigms that reflect the milieu of gene × environment and gene × gene interactions that characterize psychotic illness and its response to antipsychotic drugs.


Subject(s)
Antipsychotic Agents/therapeutic use , Disease Models, Animal , Psychotic Disorders/drug therapy , Animals , Carrier Proteins/genetics , Dopamine/physiology , Dysbindin , Dystrophin-Associated Proteins , Glutamine/physiology , Mice , Nerve Tissue Proteins/genetics , Receptors, N-Methyl-D-Aspartate/physiology , Schizophrenia/genetics , Synaptic Transmission
17.
CNS Neurol Disord Drug Targets ; 11(3): 282-91, 2012 May.
Article in English | MEDLINE | ID: mdl-22483298

ABSTRACT

Current antipsychotic drugs lack material efficacy against the negative symptoms and cognitive deficits of schizophrenia. There is considerable uncertainty regarding the optimal pharmacotherapeutic strategy for treating these and other aspects of psychotic illness. The present review summarises clinical, mutant, and psychopharmacological data related to catechol-O-methyltransferase (COMT), an enzyme involved in the catabolism of catecholamine neurotransmitters, with a view to establishing the antipsychotic potential of compounds targeting the action of this enzyme. The review examines clinical and preclinical genetic data linking COMT gene variation with risk for schizophrenia or specific symptoms or disease endophenotypes. We then summarise data concerning the behavioural effects of COMT inhibitors. These genetic and pharmacological data relating to COMT as a therapeutic target have implications for the development of individualised treatments for treatment-resistant symptoms of schizophrenia, including cognitive dysfunction and, potentially, negative symptoms.


Subject(s)
Antipsychotic Agents/therapeutic use , Brain/enzymology , Catechol O-Methyltransferase Inhibitors , Enzyme Inhibitors/therapeutic use , Schizophrenia/drug therapy , Schizophrenia/enzymology , Antipsychotic Agents/pharmacology , Brain/drug effects , Catechol O-Methyltransferase/metabolism , Enzyme Inhibitors/pharmacology , Humans
18.
Brain Behav Immun ; 26(4): 660-71, 2012 May.
Article in English | MEDLINE | ID: mdl-22426432

ABSTRACT

There is a paucity of animal models by which the contributions of environmental and genetic factors to the pathobiology of psychosis can be investigated. This study examined the individual and combined effects of chronic social stress during adolescence and deletion of the schizophrenia risk gene neuregulin-1 (NRG1) on adult mouse phenotype. Mice were exposed to repeated social defeat stress during adolescence and assessed for exploratory behaviour, working memory, sucrose preference, social behaviour and prepulse inhibition in adulthood. Thereafter, in vitro cytokine responses to mitogen stimulation and corticosterone inhibition were assayed in spleen cells, with measurement of cytokine and brain-derived neurotrophic factor (BDNF) mRNA in frontal cortex, hippocampus and striatum. NRG1 mutants exhibited hyperactivity, decreased anxiety, impaired sensorimotor gating and reduced preference for social novelty. The effects of stress on exploratory/anxiety-related parameters, spatial working memory, sucrose preference and basal cytokine levels were modified by NRG1 deletion. Stress also exerted varied effect on spleen cytokine response to concanavalin A and brain cytokine and BDNF mRNA expression in NRG1 mutants. The experience of psychosocial stress during adolescence may trigger further pathobiological features that contribute to the development of schizophrenia, particularly in those with underlying NRG1 gene abnormalities. This model elaborates the importance of gene × environment interactions in the etiology of schizophrenia.


Subject(s)
Gene-Environment Interaction , Neuregulin-1/genetics , Schizophrenia/genetics , Schizophrenic Psychology , Stress, Psychological/genetics , Animals , Brain-Derived Neurotrophic Factor/analysis , Corticosterone/analysis , Cytokines/analysis , Disease Models, Animal , Environment , Exploratory Behavior , Genotype , Mice , Mice, Knockout , Phenotype , Social Behavior
19.
Curr Top Behav Neurosci ; 12: 209-50, 2012.
Article in English | MEDLINE | ID: mdl-22367925

ABSTRACT

Schizophrenia is characterised by a multifactorial aetiology that involves genetic liability interacting with epigenetic and environmental factors to increase risk for developing the disorder. A consensus view is that the genetic component involves several common risk alleles of small effect and/or rare but penetrant copy number variations. Furthermore, there is increasing evidence for broader, overlapping genetic-phenotypic relationships in psychosis; for example, the same susceptibility genes also confer risk for bipolar disorder. Phenotypic characterisation of genetic models of candidate risk genes and/or putative pathophysiological processes implicated in schizophrenia, as well as examination of epidemiologically relevant gene × environment interactions in these models, can illuminate molecular and pathobiological mechanisms involved in schizophrenia. The present chapter outlines both the evidence from phenotypic studies in mutant mouse models related to schizophrenia and recently described mutant models addressing such gene × environment interactions. Emphasis is placed on evaluating the extent to which mutant phenotypes recapitulate the totality of the disease phenotype or model selective endophenotypes. We also discuss new developments and trends in relation to the functional genomics of psychosis which might help to inform on the construct validity of mutant models of schizophrenia and highlight methodological challenges in phenotypic evaluation that relate to such models.


Subject(s)
DNA Copy Number Variations/genetics , Endophenotypes , Genetic Predisposition to Disease , Schizophrenia/genetics , Animals , Disease Models, Animal , Humans , Schizophrenic Psychology
20.
Behav Brain Res ; 228(2): 351-8, 2012 Mar 17.
Article in English | MEDLINE | ID: mdl-22192380

ABSTRACT

Catechol-O-methyltransferase, an enzyme involved in regulating brain catecholamine levels, has been implicated in anxiety, pain and/or stress responsivity. Elements of this putative association remain unclarified, notably whether: (a) COMT variation modulates responses to acute and/or chronic stress equally; (b) acute pharmacological inhibition of COMT produces comparable effects on anxiety to that observed after deletion of the COMT gene; (c) COMT genotype modulates action of anxiolytic drugs. We aimed to further investigate the relationship between reduced COMT function, anxiety and stress responsivity in mice. To compare the effect of acute vs. chronic restraint stress in female COMT KO vs. WT mice, serum corticosterone and cytokine concentrations were measured [Experiment 1]. Sensitivity to the benzodiazepines midazolam and chlordiazepoxide in the light-dark test was assessed in female COMT KO vs. WT mice [Experiment 2]. Effects of acute administration of the COMT inhibitor tolcapone, and of these same benzodiazepines thereon, in the light-dark test were assessed in female C57BL/6 mice [Experiment 3]. COMT KO mice demonstrated an increased corticosterone response to acute but not chronic stress, and a modified cytokine profile after chronic, but not acute stress. COMT KO mice showed increased anxiety, but benzodiazepine sensitivity was affected by COMT genotype. Whilst tolcapone had no effect on light/dark performance in C57BL6/J mice it decreased benzodiazepine sensitivity. These data elaborate earlier findings of increased anxiety in female COMT KO mice and also clarify a role for COMT in modulating stress-related hormonal and immune parameters in a manner that depends on chronicity of the stressor.


Subject(s)
Anti-Anxiety Agents/therapeutic use , Catechol O-Methyltransferase/deficiency , Chlordiazepoxide/therapeutic use , Midazolam/therapeutic use , Stress, Psychological , Analysis of Variance , Animals , Behavior, Animal/physiology , Benzophenones/therapeutic use , Corticosterone/blood , Cytokines/blood , Dark Adaptation/drug effects , Dark Adaptation/genetics , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/therapeutic use , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitrophenols/therapeutic use , Organ Size/drug effects , Restraint, Physical/adverse effects , Statistics as Topic , Stress, Psychological/drug therapy , Stress, Psychological/physiopathology , Stress, Psychological/psychology , Time Factors , Tolcapone
SELECTION OF CITATIONS
SEARCH DETAIL