1
|
Xu H, Yang F. The interplay of dopamine metabolism abnormalities and mitochondrial defects in the pathogenesis of schizophrenia. Transl Psychiatry 2022; 12:464. [PMID: 36344514 PMCID: PMC9640700 DOI: 10.1038/s41398-022-02233-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 11/09/2022] Open
Abstract
Dopamine (DA) is a major monoamine neurotransmitter in the brain and has essential roles in higher functions of the brain. Malfunctions of dopaminergic signaling have been implicated in various mental disorders such as addiction, attention deficit/hyperactivity disorder, Huntington's disease, Parkinson's disease (PD), and schizophrenia. The pathogenesis of PD and schizophrenia involves the interplay of mitochondrial defect and DA metabolism abnormalities. This article focuses on this issue in schizophrenia. It started with the introduction of metabolism, behavioral action, and physiology of DA, followed by reviewing evidence for malfunctions of dopaminergic signaling in patients with schizophrenia. Then it provided an overview of multiple facets of mitochondrial physiology before summarizing mitochondrial defects reported in clinical studies with schizophrenia patients. Finally, it discussed the interplay between DA metabolism abnormalities and mitochondrial defects and outlined some clinical studies showing effects of combination therapy of antipsychotics and antioxidants in treating patients with schizophrenia. The update and integration of these lines of information may advance our understanding of the etiology, pathogenesis, phenomenology, and treatment of schizophrenia.
Collapse
Affiliation(s)
- Haiyun Xu
- School of Mental Health, Wenzhou Medical University, Wenzhou, China.
- Zhejiang Provincial Clinical Research Center for Mental Illness, The Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, China.
- Mental Health Center, Shantou University Medical College, Shantou, China.
| | - Fan Yang
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
2
|
Guo L, Xiao P, Zhang X, Yang Y, Yang M, Wang T, Lu H, Tian H, Wang H, Liu J. Inulin ameliorates schizophrenia via modulation of the gut microbiota and anti-inflammation in mice. Food Funct 2021; 12:1156-1175. [PMID: 33432310 DOI: 10.1039/d0fo02778b] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The microbiome-gut-brain (MGB) axis, which regulates neurological and cognitive functions, plays an essential role in schizophrenia (SCZ) progression. Dietary inulin could be a novel strategy for the treatment of SCZ due to its modulating effects on the gut microbiota. In this study, the effects of inulin on mice with SCZ were studied. As indicated by the behavioural tests, expression of neurotransmitters, inflammatory indicators, and brain morphology, inulin administration ameliorated aberrant behaviours (locomotor hypoactivity, anxiety disorders and depressive behaviours, and impaired learning and spatial recognition memory) and effectively reduced neuroinflammation and neuronal damage. In addition, inulin improved intestinal integrity and permeability, as indicated by the elevated expression of tight junction proteins (p < 0.05). The results of 16S rRNA sequencing and analysis showed that inulin increased the abundance of Lactobacillus and Bifidobacterium, which were negatively correlated with 5-hydroxytryptamine and inflammatory cytokines and positively correlated with brain-derived neurotrophic factor (BDNF). Inulin caused a reduction in Akkermansia that was positively correlated with inflammatory cytokines and negatively correlated with BDNF. These results suggested that dietary inulin modulated the gut microbiota and exerted anti-inflammatory effects in mice though the MGB axis, which further ameliorated SCZ. Therefore, the results of this study provide a potential explanation for inulin intervention in the treatment of SCZ.
Collapse
Affiliation(s)
- Li Guo
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Peilun Xiao
- Department of Anatomy, Weifang Medical University, Weifang 261042, Shandong, China.
| | - Xiaoxia Zhang
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Yang Yang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Miao Yang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Ting Wang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Haixia Lu
- Clinical Medical College, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Hongyan Tian
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Hao Wang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Juan Liu
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| |
Collapse
|
3
|
Kokkinou M, Irvine EE, Bonsall DR, Natesan S, Wells LA, Smith M, Glegola J, Paul EJ, Tossell K, Veronese M, Khadayate S, Dedic N, Hopkins SC, Ungless MA, Withers DJ, Howes OD. Reproducing the dopamine pathophysiology of schizophrenia and approaches to ameliorate it: a translational imaging study with ketamine. Mol Psychiatry 2021; 26:2562-2576. [PMID: 32382134 PMCID: PMC8440182 DOI: 10.1038/s41380-020-0740-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 04/06/2020] [Accepted: 04/17/2020] [Indexed: 12/14/2022]
Abstract
Patients with schizophrenia show increased striatal dopamine synthesis capacity in imaging studies. The mechanism underlying this is unclear but may be due to N-methyl-D-aspartate receptor (NMDAR) hypofunction and parvalbumin (PV) neuronal dysfunction leading to disinhibition of mesostriatal dopamine neurons. Here, we develop a translational mouse model of the dopamine pathophysiology seen in schizophrenia and test approaches to reverse the dopamine changes. Mice were treated with sub-chronic ketamine (30 mg/kg) or saline and then received in vivo positron emission tomography of striatal dopamine synthesis capacity, analogous to measures used in patients. Locomotor activity was measured using the open-field test. In vivo cell-type-specific chemogenetic approaches and pharmacological interventions were used to manipulate neuronal excitability. Immunohistochemistry and RNA sequencing were used to investigate molecular mechanisms. Sub-chronic ketamine increased striatal dopamine synthesis capacity (Cohen's d = 2.5) and locomotor activity. These effects were countered by inhibition of midbrain dopamine neurons, and by activation of PV interneurons in pre-limbic cortex and ventral subiculum of the hippocampus. Sub-chronic ketamine reduced PV expression in these cortical and hippocampal regions. Pharmacological intervention with SEP-363856, a novel psychotropic agent with agonism at trace amine receptor 1 (TAAR1) and 5-HT1A receptors but no appreciable action at dopamine D2 receptors, significantly reduced the ketamine-induced increase in dopamine synthesis capacity. These results show that sub-chronic ketamine treatment in mice mimics the dopaminergic alterations in patients with psychosis, that this requires activation of midbrain dopamine neurons, and can be ameliorated by activating PV interneurons and by a TAAR1/5-HT1A agonist. This identifies novel therapeutic approaches for targeting presynaptic dopamine dysfunction in patients with schizophrenia and effects of ketamine relevant to its therapeutic use for treating major depression.
Collapse
Affiliation(s)
- Michelle Kokkinou
- MRC London Institute of Medical Sciences (LMS), London, W12 0NN, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Elaine E Irvine
- MRC London Institute of Medical Sciences (LMS), London, W12 0NN, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - David R Bonsall
- Invicro, Burlington Danes, Hammersmith Hospital, London, W12 0NN, UK
| | - Sridhar Natesan
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Lisa A Wells
- Invicro, Burlington Danes, Hammersmith Hospital, London, W12 0NN, UK
| | - Mark Smith
- MRC London Institute of Medical Sciences (LMS), London, W12 0NN, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Justyna Glegola
- MRC London Institute of Medical Sciences (LMS), London, W12 0NN, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Eleanor J Paul
- MRC London Institute of Medical Sciences (LMS), London, W12 0NN, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Kyoko Tossell
- MRC London Institute of Medical Sciences (LMS), London, W12 0NN, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Sanjay Khadayate
- MRC London Institute of Medical Sciences (LMS), London, W12 0NN, UK
| | - Nina Dedic
- Sunovion Pharmaceuticals, 84 Waterford Drive, Marlborough, MA, 01752, USA
| | - Seth C Hopkins
- Sunovion Pharmaceuticals, 84 Waterford Drive, Marlborough, MA, 01752, USA
| | - Mark A Ungless
- MRC London Institute of Medical Sciences (LMS), London, W12 0NN, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Dominic J Withers
- MRC London Institute of Medical Sciences (LMS), London, W12 0NN, UK.
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, W12 0NN, UK.
| | - Oliver D Howes
- MRC London Institute of Medical Sciences (LMS), London, W12 0NN, UK.
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, W12 0NN, UK.
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK.
| |
Collapse
|
4
|
Transplantation of microbiota from drug-free patients with schizophrenia causes schizophrenia-like abnormal behaviors and dysregulated kynurenine metabolism in mice. Mol Psychiatry 2020; 25:2905-2918. [PMID: 31391545 DOI: 10.1038/s41380-019-0475-4] [Citation(s) in RCA: 174] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/28/2019] [Accepted: 06/20/2019] [Indexed: 12/18/2022]
Abstract
Accumulating evidence suggests that gut microbiota plays a role in the pathogenesis of schizophrenia via the microbiota-gut-brain axis. This study sought to investigate whether transplantation of fecal microbiota from drug-free patients with schizophrenia into specific pathogen-free mice could cause schizophrenia-like behavioral abnormalities. The results revealed that transplantation of fecal microbiota from schizophrenic patients into antibiotic-treated mice caused behavioral abnormalities such as psychomotor hyperactivity, impaired learning and memory in the recipient animals. These mice also showed elevation of the kynurenine-kynurenic acid pathway of tryptophan degradation in both periphery and brain, as well as increased basal extracellular dopamine in prefrontal cortex and 5-hydroxytryptamine in hippocampus, compared with their counterparts receiving feces from healthy controls. Furthermore, colonic luminal filtrates from the mice transplanted with patients' fecal microbiota increased both kynurenic acid synthesis and kynurenine aminotransferase II activity in cultured hepatocytes and forebrain cortical slices. Sixty species of donor-derived bacteria showed significant difference between the mice colonized with the patients' and the controls' fecal microbiota, highlighting 78 differentially enriched functional modules including tryptophan biosynthesis function. In conclusion, our study suggests that the abnormalities in the composition of gut microbiota contribute to the pathogenesis of schizophrenia partially through the manipulation of tryptophan-kynurenine metabolism.
Collapse
|
5
|
Casamassa A, Ferrari D, Gelati M, Carella M, Vescovi AL, Rosati J. A Link between Genetic Disorders and Cellular Impairment, Using Human Induced Pluripotent Stem Cells to Reveal the Functional Consequences of Copy Number Variations in the Central Nervous System-A Close Look at Chromosome 15. Int J Mol Sci 2020; 21:ijms21051860. [PMID: 32182809 PMCID: PMC7084702 DOI: 10.3390/ijms21051860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/02/2020] [Accepted: 03/05/2020] [Indexed: 12/28/2022] Open
Abstract
Recent cutting-edge human genetics technology has allowed us to identify copy number variations (CNVs) and has provided new insights for understanding causative mechanisms of human diseases. A growing number of studies show that CNVs could be associated with physiological mechanisms linked to evolutionary trigger, as well as to the pathogenesis of various diseases, including cancer, autoimmune disease and mental disorders such as autism spectrum disorders, schizophrenia, intellectual disabilities or attention-deficit/hyperactivity disorder. Their incomplete penetrance and variable expressivity make diagnosis difficult and hinder comprehension of the mechanistic bases of these disorders. Additional elements such as co-presence of other CNVs, genomic background and environmental factors are involved in determining the final phenotype associated with a CNV. Genetically engineered animal models are helpful tools for understanding the behavioral consequences of CNVs. However, the genetic background and the biology of these animal model systems have sometimes led to confusing results. New cellular models obtained through somatic cellular reprogramming technology that produce induced pluripotent stem cells (iPSCs) from human subjects are being used to explore the mechanisms involved in the pathogenic consequences of CNVs. Considering the vast quantity of CNVs found in the human genome, we intend to focus on reviewing the current literature on the use of iPSCs carrying CNVs on chromosome 15, highlighting advantages and limits of this system with respect to mouse model systems.
Collapse
Affiliation(s)
- Alessia Casamassa
- Cellular Reprogramming Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini 1, 71013 San Giovanni Rotondo, Foggia, Italy;
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, Viale Abramo Lincoln 5, 81100 Caserta, Italy
| | - Daniela Ferrari
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy;
| | - Maurizio Gelati
- Fondazione IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini 1, 71013 San Giovanni Rotondo, Foggia, Italy; (M.G.); (M.C.)
| | - Massimo Carella
- Fondazione IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini 1, 71013 San Giovanni Rotondo, Foggia, Italy; (M.G.); (M.C.)
| | - Angelo Luigi Vescovi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy;
- Fondazione IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini 1, 71013 San Giovanni Rotondo, Foggia, Italy; (M.G.); (M.C.)
- Correspondence: (A.L.V.); (J.R.)
| | - Jessica Rosati
- Cellular Reprogramming Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini 1, 71013 San Giovanni Rotondo, Foggia, Italy;
- Correspondence: (A.L.V.); (J.R.)
| |
Collapse
|
6
|
Waddington JL, Zhen X, O'Tuathaigh CMP. Developmental Genes and Regulatory Proteins, Domains of Cognitive Impairment in Schizophrenia Spectrum Psychosis and Implications for Antipsychotic Drug Discovery: The Example of Dysbindin-1 Isoforms and Beyond. Front Pharmacol 2020; 10:1638. [PMID: 32063853 PMCID: PMC7000454 DOI: 10.3389/fphar.2019.01638] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 12/16/2019] [Indexed: 12/19/2022] Open
Abstract
Alongside positive and negative symptomatology, deficits in working memory, attention, selective learning processes, and executive function have been widely documented in schizophrenia spectrum psychosis. These cognitive abnormalities are strongly associated with impairment across multiple function domains and are generally treatment-resistant. The DTNBP1 (dystrobrevin-binding protein-1) gene, encoding dysbindin, is considered a risk factor for schizophrenia and is associated with variation in cognitive function in both clinical and nonclinical samples. Downregulation of DTNBP1 expression in dorsolateral prefrontal cortex and hippocampal formation of patients with schizophrenia has been suggested to serve as a primary pathophysiological process. Described as a "hub," dysbindin is an important regulatory protein that is linked with multiple complexes in the brain and is involved in a wide variety of functions implicated in neurodevelopment and neuroplasticity. The expression pattern of the various dysbindin isoforms (-1A, -1B, -1C) changes depending upon stage of brain development, tissue areas and subcellular localizations, and can involve interaction with different protein partners. We review evidence describing how sequence variation in DTNBP1 isoforms has been differentially associated with schizophrenia-associated symptoms. We discuss results linking these isoform proteins, and their interacting molecular partners, with cognitive dysfunction in schizophrenia, including evidence from drosophila through to genetic mouse models of dysbindin function. Finally, we discuss preclinical evidence investigating the antipsychotic potential of molecules that influence dysbindin expression and functionality. These studies, and other recent work that has extended this approach to other developmental regulators, may facilitate identification of novel molecular pathways leading to improved antipsychotic treatments.
Collapse
Affiliation(s)
- John L Waddington
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland.,Jiangsu Key Laboratory of Translational Research & Therapy for Neuro-Psychiatric Disorders and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Xuechu Zhen
- Jiangsu Key Laboratory of Translational Research & Therapy for Neuro-Psychiatric Disorders and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Colm M P O'Tuathaigh
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland.,Medical Education Unit, School of Medicine, Brookfield Health Sciences Complex, University College Cork, Cork, Ireland
| |
Collapse
|
7
|
Demin KA, Meshalkina DA, Volgin AD, Yakovlev OV, de Abreu MS, Alekseeva PA, Friend AJ, Lakstygal AM, Zabegalov K, Amstislavskaya TG, Strekalova T, Bao W, Kalueff AV. Developing zebrafish experimental animal models relevant to schizophrenia. Neurosci Biobehav Rev 2019; 105:126-133. [DOI: 10.1016/j.neubiorev.2019.07.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 07/20/2019] [Accepted: 07/27/2019] [Indexed: 12/18/2022]
|
8
|
Wang P, Cao T, Chen J, Jiang Y, Wang C, Waddington JL, Zhen X. D2 receptor-mediated miRNA-143 expression is associated with the effects of antipsychotic drugs on phencyclidine-induced schizophrenia-related locomotor hyperactivity and with Neuregulin-1 expression in mice. Neuropharmacology 2019; 157:107675. [DOI: 10.1016/j.neuropharm.2019.107675] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 05/09/2019] [Accepted: 06/17/2019] [Indexed: 12/21/2022]
|
9
|
Bolker JA. Selection of Models: Evolution and the Choice of Species for Translational Research. BRAIN, BEHAVIOR AND EVOLUTION 2019; 93:82-91. [PMID: 31416088 DOI: 10.1159/000500317] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 04/10/2019] [Indexed: 11/19/2022]
Abstract
Evolutionary thinking can inform the choice and assessment of model species in neuroscience, particularly when such models are intended to generate knowledge that will translate to humans. Avoiding errors that arise from oversimplified notions of phylogeny or genotype-phenotype mapping is one contribution; evolutionary biology also offers positive guidance. The challenge of finding adequate non-human models for translational research is particularly acute in neuroscience: neurobiological and behavioral phenotypes are complex and plastic, and many traits important in humans are absent, radically different, or difficult to assess in other species. Evolutionary perspectives help to articulate and address these challenges. Darwin's description of "descent with modification" points to two aspects of evolution that can help us assess the matching between a prospective model species and its intended target. One is trees that represent the structure of phylogenetic relationships; the other is phenotypic traits, i.e. the unique characteristics of each species' evolved biology and natural history. Mapping traits onto a phylogeny is the first step toward analyzing the source of similarities between a target and a potential model. Whether similar traits arise from shared ancestry or from adaptive convergence has important implications for what kinds of inferences can be justified, and for the likely translatability of findings. Evolution offers both a rich source of possible models, and guidance for choosing the best ones for a given purpose. Considering model choice from an evolutionary angle not only helps to answer the question "What species might be a good model for studying x?" but also suggests additional questions we should be asking to assess the utility of both potential and current models. Recognizing the diverse ways model organisms can function expands our search image as we seek species to study that can both extend general knowledge, and generate translatable insights relevant to human neurobiology and disease.
Collapse
Affiliation(s)
- Jessica A Bolker
- Department of Biological Sciences, University of New Hampshire, Durham, New Hampshire, USA,
| |
Collapse
|
10
|
Abstract
Hippocampal abnormalities have been heavily implicated in the pathophysiology of schizophrenia. The dentate gyrus of the hippocampus was shown to manifest an immature molecular profile in schizophrenia subjects, as well as in various animal models of the disorder. In this position paper, we advance a hypothesis that this immature molecular profile is accompanied by an identifiable immature morphology of the dentate gyrus granule cell layer. We adduce evidence for arrested maturation of the dentate gyrus in the human schizophrenia-affected brain, as well as multiple rodent models of the disease. Implications of this neurohistopathological signature for current theory regarding the development of schizophrenia are discussed.
Collapse
Affiliation(s)
- Ayda Tavitian
- Department of Neurology & Neurosurgery, Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Wei Song
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Hyman M. Schipper
- Department of Neurology & Neurosurgery, Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
11
|
Winship IR, Dursun SM, Baker GB, Balista PA, Kandratavicius L, Maia-de-Oliveira JP, Hallak J, Howland JG. An Overview of Animal Models Related to Schizophrenia. CANADIAN JOURNAL OF PSYCHIATRY. REVUE CANADIENNE DE PSYCHIATRIE 2019; 64:5-17. [PMID: 29742910 PMCID: PMC6364139 DOI: 10.1177/0706743718773728] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
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.
Collapse
Affiliation(s)
- Ian R Winship
- 1 Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta
| | - Serdar M Dursun
- 2 Department of Psychiatry, Neurochemical Research Unit and Bebensee Schizophrenia Research Unit, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta.,3 National Institute of Science and Technology-Translational Science, Brazil
| | - Glen B Baker
- 2 Department of Psychiatry, Neurochemical Research Unit and Bebensee Schizophrenia Research Unit, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta.,3 National Institute of Science and Technology-Translational Science, Brazil
| | - Priscila A Balista
- 4 Department of Pharmacy, Centro Universitario das Faculdades Metropolitanas Unidas, São Paulo, Brazil
| | - Ludmyla Kandratavicius
- 5 Department of Neuroscience and Behavior, Faculty of Medicine of Ribeirao Preto, University of São Paulo, Ribeirao Preto, Brazil
| | - Joao Paulo Maia-de-Oliveira
- 3 National Institute of Science and Technology-Translational Science, Brazil.,6 Department of Clinical Medicine, Rio Grande do Norte Federal University, Natal, Brazil
| | - Jaime Hallak
- 3 National Institute of Science and Technology-Translational Science, Brazil.,5 Department of Neuroscience and Behavior, Faculty of Medicine of Ribeirao Preto, University of São Paulo, Ribeirao Preto, Brazil.,7 Department of Psychiatry (NRU), University of Alberta, Edmonton, Alberta
| | - John G Howland
- 8 Department of Physiology, University of Saskatchewan, Saskatoon, Saskatchewan
| |
Collapse
|
12
|
Waddington JL, O'Tuathaigh CM. Modelling the neuromotor abnormalities of psychotic illness: Putative mechanisms and systems dysfunction. Schizophr Res 2018; 200:12-19. [PMID: 28867516 DOI: 10.1016/j.schres.2017.08.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/11/2017] [Accepted: 08/16/2017] [Indexed: 12/20/2022]
Abstract
Limitations in access to antipsychotic-naïve patients and in the incisiveness of studies that can be conducted on them, together with the inevitability of subsequent antipsychotic treatment, indicate an enduring role for animal models that can inform on the pathobiology of neuromotor abnormalities in schizophrenia and related psychotic illness. This review focusses particularly on genetically modified mouse models that involve genes associated with risk for schizophrenia and with mechanisms implicated in the neuromotor abnormalities evident in psychotic patients, as well as developmental models that seek to mirror the trajectory, phenomenology and putative pathophysiology of psychotic illness. Such abnormalities are inconsistent and subtle in mice mutant for some schizophrenia risk genes but more evident for others. The phenotype of dopaminergic and glutamatergic mutants indicates the involvement of these mechanisms, informs on the roles of specific receptor subtypes, and implicates the interplay of cortical and subcortical processes. Developmental models suggest a criticality in the timing of early adversity for diversity in the relative emergence of psychological symptoms vis-à-vis neuromotor abnormalities in the overall psychosis phenotype. These findings elaborate current concepts of dysfunction in a neuronal network linking the cerebral cortex, basal ganglia, thalamus and cerebellum. Both findings in model systems and clinical evidence converge in indicating that any distinction between 'psychomotor' and 'neuromotor' abnormality is artificial and arbitrary due to a unitary origin in developmentally determined systems/network dysfunction that underlies the lifetime trajectory of psychotic illness.
Collapse
Affiliation(s)
- John L Waddington
- Molecular & Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland; Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psychiatric-Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China.
| | | |
Collapse
|
13
|
Khan A, Powell SB. Sensorimotor gating deficits in "two-hit" models of schizophrenia risk factors. Schizophr Res 2018; 198:68-83. [PMID: 29070440 PMCID: PMC5911431 DOI: 10.1016/j.schres.2017.10.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 10/03/2017] [Accepted: 10/06/2017] [Indexed: 02/07/2023]
Abstract
Genetic and environmental models of neuropsychiatric disease have grown exponentially over the last 20years. One measure that is often used to evaluate the translational relevance of these models to human neuropsychiatric disease is prepulse inhibition of startle (PPI), an operational measure of sensorimotor gating. Deficient PPI characterizes several neuropsychiatric disorders but has been most extensively studied in schizophrenia. It has become a useful tool in translational neuropharmacological and molecular genetics studies because it can be measured across species using almost the same experimental parameters. Although initial studies of PPI in rodents were pharmacological because of the robust predictive validity of PPI for antipsychotic efficacy, more recently, PPI has become standard common behavioral measures used in genetic and neurodevelopmental models of schizophrenia. Here we review "two hit" models of schizophrenia and discuss the utility of PPI as a tool in phenotyping these models of relevant risk factors. In the review, we consider approaches to rodent models of genetic and neurodevelopmental risk factors and selectively review "two hit" models of gene×environment and environment×environment interactions in which PPI has been measured.
Collapse
Affiliation(s)
- Asma Khan
- Department of Psychiatry, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, United States; Research Service, VA San Diego Healthcare System, La Jolla, CA, United States
| | - Susan B Powell
- Department of Psychiatry, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, United States; Research Service, VA San Diego Healthcare System, La Jolla, CA, United States.
| |
Collapse
|
14
|
Kesby JP, Eyles DW, McGrath JJ, Scott JG. Dopamine, psychosis and schizophrenia: the widening gap between basic and clinical neuroscience. Transl Psychiatry 2018; 8:30. [PMID: 29382821 PMCID: PMC5802623 DOI: 10.1038/s41398-017-0071-9] [Citation(s) in RCA: 205] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/10/2017] [Accepted: 10/26/2017] [Indexed: 12/18/2022] Open
Abstract
The stagnation in drug development for schizophrenia highlights the need for better translation between basic and clinical research. Understanding the neurobiology of schizophrenia presents substantial challenges but a key feature continues to be the involvement of subcortical dopaminergic dysfunction in those with psychotic symptoms. Our contemporary knowledge regarding dopamine dysfunction has clarified where and when dopaminergic alterations may present in schizophrenia. For example, clinical studies have shown patients with schizophrenia show increased presynaptic dopamine function in the associative striatum, rather than the limbic striatum as previously presumed. Furthermore, subjects deemed at high risk of developing schizophrenia show similar presynaptic dopamine abnormalities in the associative striatum. Thus, our view of subcortical dopamine function in schizophrenia continues to evolve as we accommodate this newly acquired information. However, basic research in animal models has been slow to incorporate these clinical findings. For example, psychostimulant-induced locomotion, the commonly utilised phenotype for positive symptoms in rodents, is heavily associated with dopaminergic activation in the limbic striatum. This anatomical misalignment has brought into question how we assess positive symptoms in animal models and represents an opportunity for improved translation between basic and clinical research. The current review focuses on the role of subcortical dopamine dysfunction in psychosis and schizophrenia. We present and discuss alternative phenotypes that may provide a more translational approach to assess the neurobiology of positive symptoms in schizophrenia. Incorporation of recent clinical findings is essential if we are to develop meaningful translational animal models.
Collapse
Affiliation(s)
- JP Kesby
- 0000 0000 9320 7537grid.1003.2Queensland Brain Institute, The University of Queensland, St. Lucia, QLD Australia ,0000 0000 9320 7537grid.1003.2Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD Australia
| | - DW Eyles
- 0000 0000 9320 7537grid.1003.2Queensland Brain Institute, The University of Queensland, St. Lucia, QLD Australia ,0000 0004 0606 3563grid.417162.7Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, QLD Australia
| | - JJ McGrath
- 0000 0000 9320 7537grid.1003.2Queensland Brain Institute, The University of Queensland, St. Lucia, QLD Australia ,0000 0004 0606 3563grid.417162.7Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, QLD Australia ,0000 0001 1956 2722grid.7048.bNational Centre for Register-based Research, Aarhus University, Aarhus C, Denmark
| | - JG Scott
- 0000 0000 9320 7537grid.1003.2Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD Australia ,0000 0004 0606 3563grid.417162.7Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, QLD Australia ,0000 0001 0688 4634grid.416100.2Metro North Mental Health, Royal Brisbane and Women’s Hospital, Herston, QLD Australia
| |
Collapse
|
15
|
Waddington JL, Katina S, O'Tuathaigh CMP, Bowman AW. Translational Genetic Modelling of 3D Craniofacial Dysmorphology: Elaborating the Facial Phenotype of Neurodevelopmental Disorders Through the "Prism" of Schizophrenia. Curr Behav Neurosci Rep 2017; 4:322-330. [PMID: 29201594 PMCID: PMC5694503 DOI: 10.1007/s40473-017-0136-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Purpose of Review In the context of human developmental conditions, we review the conceptualisation of schizophrenia as a neurodevelopmental disorder, the status of craniofacial dysmorphology as a clinically accessible index of brain dysmorphogenesis, the ability of genetically modified mouse models of craniofacial dysmorphology to inform on the underlying dysmorphogenic process and how geometric morphometric techniques in mutant mice can extend quantitative analysis. Recent Findings Mutant mice with disruption of neuregulin-1, a gene associated meta-analytically with risk for schizophrenia, constitute proof-of-concept studies of murine facial dysmorphology in a manner analogous to clinical studies in schizophrenia. Geometric morphometric techniques informed on the topography of facial dysmorphology and identified asymmetry therein. Summary Targeted disruption in mice of genes involved in individual components of developmental processes and analysis of resultant facial dysmorphology using geometric morphometrics can inform on mechanisms of dysmorphogenesis at levels of incisiveness not possible in human subjects.
Collapse
Affiliation(s)
- John L Waddington
- Molecular & Cellular Therapeutics, Royal College of Surgeons in Ireland, St. Stephen's Green, Dublin 2, Ireland.,Jiangsu Key Laboratory of Translational Research & Therapy for Neuro-Psychiatric-Disorders and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123 China
| | - Stanislav Katina
- School of Mathematics and Statistics, University of Glasgow, Glasgow, G12 8QQ UK.,Institute of Mathematics and Statistics, Masaryk University, Brno, Czech Republic.,Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, Bratislava, Slovakia
| | | | - Adrian W Bowman
- School of Mathematics and Statistics, University of Glasgow, Glasgow, G12 8QQ UK
| |
Collapse
|
16
|
Bolker JA. Animal Models in Translational Research: Rosetta Stone or Stumbling Block? Bioessays 2017; 39. [PMID: 29052843 DOI: 10.1002/bies.201700089] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 09/17/2017] [Indexed: 01/12/2023]
Abstract
Leading animal models are powerful tools for translational research, but they also present obstacles. Poorly conducted preclinical research in animals is a common cause of translational failure, but even when such research is well-designed and carefully executed, challenges remain. In particular, dominant models may bias research directions, elide essential aspects of human disease, omit important context, or subtly shift research targets. Recognizing these stumbling blocks can help us find ways to avoid them: employing a wider range of models, incorporating more realistic environmental conditions, better aligning studies between animals and patients, and focusing on human biology and therapeutic goals. Such changes are costly; but insisting it would be impractical or unrealistic to change strategies offers no way out of the current impasse. Rather, we must acknowledge the obstacles as well as the advantages presented by core models, and direct some of our investments in translational research toward getting around them.
Collapse
Affiliation(s)
- Jessica A Bolker
- Department of Biological Sciences, University of New Hampshire, Durham, NH, USA
| |
Collapse
|
17
|
O’Tuathaigh CMP, Mathur N, O’Callaghan MJ, MacIntyre L, Harvey R, Lai D, Waddington JL, Pickard BS, Watson DG, Moran PM. Specialized Information Processing Deficits and Distinct Metabolomic Profiles Following TM-Domain Disruption of Nrg1. Schizophr Bull 2017; 43:1100-1113. [PMID: 28338897 PMCID: PMC5581893 DOI: 10.1093/schbul/sbw189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Although there is considerable genetic and pathologic evidence for an association between neuregulin 1 (NRG1) dysregulation and schizophrenia, the underlying molecular and cellular mechanisms remain unclear. Mutant mice containing disruption of the transmembrane (TM) domain of the NRG1 gene constitute a heuristic model for dysregulation of NRG1-ErbB4 signaling in schizophrenia. The present study focused on hitherto uncharacterized information processing phenotypes in this mutant line. Using a mass spectrometry-based metabolomics approach, we also quantified levels of unique metabolites in brain. Across 2 different sites and protocols, Nrg1 mutants demonstrated deficits in prepulse inhibition, a measure of sensorimotor gating, that is, disrupted in schizophrenia; these deficits were partially reversed by acute treatment with second, but not first-, generation antipsychotic drugs. However, Nrg1 mutants did not show a specific deficit in latent inhibition, a measure of selective attention that is also disrupted in schizophrenia. In contrast, in a "what-where-when" object recognition memory task, Nrg1 mutants displayed sex-specific (males only) disruption of "what-when" performance, indicative of impaired temporal aspects of episodic memory. Differential metabolomic profiling revealed that these behavioral phenotypes were accompanied, most prominently, by alterations in lipid metabolism pathways. This study is the first to associate these novel physiological mechanisms, previously independently identified as being abnormal in schizophrenia, with disruption of NRG1 function. These data suggest novel mechanisms by which compromised neuregulin function from birth might lead to schizophrenia-relevant behavioral changes in adulthood.
Collapse
Affiliation(s)
| | - Naina Mathur
- School of Psychology, University of Nottingham, Nottingham, UK
| | | | - Lynsey MacIntyre
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Richard Harvey
- Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Donna Lai
- Victor Chang Cardiac Research Institute, Sydney, Australia
| | - John L Waddington
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Jiangsu Key Laboratory of Translational Research & Therapy for Neuro-Psychiatric-Disorders and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Benjamin S Pickard
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - David G Watson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Paula M Moran
- School of Psychology, University of Nottingham, Nottingham, UK
| |
Collapse
|
18
|
O'Tuathaigh CMP, Moran PM, Zhen XC, Waddington JL. Translating advances in the molecular basis of schizophrenia into novel cognitive treatment strategies. Br J Pharmacol 2017; 174:3173-3190. [PMID: 28667666 DOI: 10.1111/bph.13938] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 06/07/2017] [Accepted: 06/12/2017] [Indexed: 02/06/2023] Open
Abstract
The presence and severity of cognitive symptoms, including working memory, executive dysfunction and attentional impairment, contributes materially to functional impairment in schizophrenia. Cognitive symptoms have proved to be resistant to both first- and second-generation antipsychotic drugs. Efforts to develop a consensus set of cognitive domains that are both disrupted in schizophrenia and are amenable to cross-species validation (e.g. the National Institute of Mental Health Cognitive Neuroscience Treatment Research to Improve Cognition in Schizophrenia and Research Domain Criteria initiatives) are an important step towards standardization of outcome measures that can be used in preclinical testing of new drugs. While causative genetic mutations have not been identified, new technologies have identified novel genes as well as hitherto candidate genes previously implicated in the pathophysiology of schizophrenia and/or mechanisms of antipsychotic efficacy. This review comprises a selective summary of these developments, particularly phenotypic data arising from preclinical genetic models for cognitive dysfunction in schizophrenia, with the aim of indicating potential new directions for pro-cognitive therapeutics. Linked Articles This article is part of a themed section on Pharmacology of Cognition: a Panacea for Neuropsychiatric Disease? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.19/issuetoc.
Collapse
Affiliation(s)
- Colm M P O'Tuathaigh
- School of Medicine, University College Cork, Brookfield Health Sciences Complex, Cork, Ireland
| | - Paula M Moran
- School of Psychology, University of Nottingham, Nottingham, UK
| | - Xuechu C Zhen
- Jiangsu Key Laboratory of Translational Research & Therapy for Neuropsychiatric Disorders and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - John L Waddington
- Jiangsu Key Laboratory of Translational Research & Therapy for Neuropsychiatric Disorders and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China.,Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| |
Collapse
|
19
|
The Schizophrenia-Associated BRD1 Gene Regulates Behavior, Neurotransmission, and Expression of Schizophrenia Risk Enriched Gene Sets in Mice. Biol Psychiatry 2017; 82:62-76. [PMID: 27837920 DOI: 10.1016/j.biopsych.2016.08.037] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 08/18/2016] [Accepted: 08/29/2016] [Indexed: 11/21/2022]
Abstract
BACKGROUND The schizophrenia-associated BRD1 gene encodes a transcriptional regulator whose comprehensive chromatin interactome is enriched with schizophrenia risk genes. However, the biology underlying the disease association of BRD1 remains speculative. METHODS This study assessed the transcriptional drive of a schizophrenia-associated BRD1 risk variant in vitro. Accordingly, to examine the effects of reduced Brd1 expression, we generated a genetically modified Brd1+/- mouse and subjected it to behavioral, electrophysiological, molecular, and integrative genomic analyses with focus on schizophrenia-relevant parameters. RESULTS Brd1+/- mice displayed cerebral histone H3K14 hypoacetylation and a broad range of behavioral changes with translational relevance to schizophrenia. These behaviors were accompanied by striatal dopamine/serotonin abnormalities and cortical excitation-inhibition imbalances involving loss of parvalbumin immunoreactive interneurons. RNA-sequencing analyses of cortical and striatal micropunches from Brd1+/- and wild-type mice revealed differential expression of genes enriched for schizophrenia risk, including several schizophrenia genome-wide association study risk genes (e.g., calcium channel subunits [Cacna1c and Cacnb2], cholinergic muscarinic receptor 4 [Chrm4)], dopamine receptor D2 [Drd2], and transcription factor 4 [Tcf4]). Integrative analyses further found differentially expressed genes to cluster in functional networks and canonical pathways associated with mental illness and molecular signaling processes (e.g., glutamatergic, monoaminergic, calcium, cyclic adenosine monophosphate [cAMP], dopamine- and cAMP-regulated neuronal phosphoprotein 32 kDa [DARPP-32], and cAMP responsive element binding protein signaling [CREB]). CONCLUSIONS Our study bridges the gap between genetic association and pathogenic effects and yields novel insights into the unfolding molecular changes in the brain of a new schizophrenia model that incorporates genetic risk at three levels: allelic, chromatin interactomic, and brain transcriptomic.
Collapse
|
20
|
Håkansson K, Runker AE, O'Sullivan GJ, Mitchell KJ, Waddington JL, O'Tuathaigh CMP. Semaphorin 6A knockout mice display abnormalities across ethologically-based topographies of exploration and in motor learning. Neurosci Lett 2017; 641:70-76. [PMID: 28109776 DOI: 10.1016/j.neulet.2017.01.043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/11/2017] [Accepted: 01/17/2017] [Indexed: 01/04/2023]
Abstract
Semaphorins are secreted or membrane-bound proteins implicated in neurodevelopmental processes of axon guidance and cell migration. Exploratory behaviour and motor learning was examined ethologically in Semaphorin 6A (Sema6A) mutant mice. The ethogram of initial exploration in Sema6A knockout mice was characterised by increased rearing to wall with decreased sifting; over subsequent habituation, locomotion, sniffing and rearing to wall were increased, with reduced habituation of rearing seated. Rotarod analysis indicated delayed motor learning in Sema6A heterozygous mutants. Disruption to the axonal guidance and cell migration processes regulated by Sema6A is associated with topographically specific disruption to fundamental aspects of behaviour, namely the ethogram of initial exploration and subsequent habituation to the environment, and motor learning.
Collapse
Affiliation(s)
- Kerstin Håkansson
- Molecular & Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Annette E Runker
- Smurfit Institute of Genetics and Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Gerard J O'Sullivan
- Molecular & Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Kevin J Mitchell
- Smurfit Institute of Genetics and Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - John L Waddington
- Molecular & Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Colm M P O'Tuathaigh
- Molecular & Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland; School of Medicine, University College Cork, Cork, Ireland.
| |
Collapse
|
21
|
Gene × Environment Interactions in Schizophrenia: Evidence from Genetic Mouse Models. Neural Plast 2016; 2016:2173748. [PMID: 27725886 PMCID: PMC5048038 DOI: 10.1155/2016/2173748] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 07/20/2016] [Accepted: 08/21/2016] [Indexed: 02/06/2023] Open
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.
Collapse
|
22
|
Sigurdsson T. Neural circuit dysfunction in schizophrenia: Insights from animal models. Neuroscience 2016; 321:42-65. [DOI: 10.1016/j.neuroscience.2015.06.059] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 06/15/2015] [Accepted: 06/26/2015] [Indexed: 12/17/2022]
|
23
|
Burrows EL, Hannan AJ. Cognitive endophenotypes, gene-environment interactions and experience-dependent plasticity in animal models of schizophrenia. Biol Psychol 2015; 116:82-9. [PMID: 26687973 DOI: 10.1016/j.biopsycho.2015.11.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Revised: 11/26/2015] [Accepted: 11/30/2015] [Indexed: 12/22/2022]
Abstract
Schizophrenia is a devastating brain disorder caused by a complex and heterogeneous combination of genetic and environmental factors. In order to develop effective new strategies to prevent and treat schizophrenia, valid animal models are required which accurately model the disorder, and ideally provide construct, face and predictive validity. The cognitive deficits in schizophrenia represent some of the most debilitating symptoms and are also currently the most poorly treated. Therefore it is crucial that animal models are able to capture the cognitive dysfunction that characterizes schizophrenia, as well as the negative and psychotic symptoms. The genomes of mice have, prior to the recent gene-editing revolution, proven the most easily manipulable of mammalian laboratory species, and hence most genetic targeting has been performed using mouse models. Importantly, when key environmental factors of relevance to schizophrenia are experimentally manipulated, dramatic changes in the phenotypes of these animal models are often observed. We will review recent studies in rodent models which provide insight into gene-environment interactions in schizophrenia. We will focus specifically on environmental factors which modulate levels of experience-dependent plasticity, including environmental enrichment, cognitive stimulation, physical activity and stress. The insights provided by this research will not only help refine the establishment of optimally valid animal models which facilitate development of novel therapeutics, but will also provide insight into the pathogenesis of schizophrenia, thus identifying molecular and cellular targets for future preclinical and clinical investigations.
Collapse
Affiliation(s)
- Emma L Burrows
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC 3010, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC 3010, Australia; Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
24
|
Habela CW, Song H, Ming GL. Modeling synaptogenesis in schizophrenia and autism using human iPSC derived neurons. Mol Cell Neurosci 2015; 73:52-62. [PMID: 26655799 DOI: 10.1016/j.mcn.2015.12.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 11/17/2015] [Accepted: 12/01/2015] [Indexed: 02/08/2023] Open
Abstract
Schizophrenia (SCZ) and autism spectrum disorder (ASD) are genetically and phenotypically complex disorders of neural development. Human genetic studies, as well as studies examining structural changes at the cellular level, have converged on glutamatergic synapse formation, function, and maintenance as common pathophysiologic substrates involved in both disorders. Synapses as basic functional units of the brain are continuously modified by experience throughout life, therefore they are particularly attractive candidates for targeted therapy. Until recently we lacked a system to evaluate dynamic changes that lead to synaptic abnormalities. With the development of techniques to generate induced pluripotent stem cells (iPSCs) from patients, we are now able to study neuronal and synaptic development in cells from individual patients in the context of genetic changes conferring disease susceptibility. In this review, we discuss recent studies focusing on neural cells differentiated from SCZ and ASD patient iPSCs. These studies support a central role for glutamatergic synapse formation and function in both disorders and demonstrate that iPSC derived neurons offer a potential system for further evaluation of processes leading to synaptic dysregulation and for the design and screening of future therapies.
Collapse
Affiliation(s)
- Christa W Habela
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Hongjun Song
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Guo-Li Ming
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
25
|
Ayhan Y, McFarland R, Pletnikov MV. Animal models of gene-environment interaction in schizophrenia: A dimensional perspective. Prog Neurobiol 2015; 136:1-27. [PMID: 26510407 DOI: 10.1016/j.pneurobio.2015.10.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 09/07/2015] [Accepted: 10/22/2015] [Indexed: 12/12/2022]
Abstract
Schizophrenia has long been considered as a disorder with multifactorial origins. Recent discoveries have advanced our understanding of the genetic architecture of the disease. However, even with the increase of identified risk variants, heritability estimates suggest an important contribution of non-genetic factors. Various environmental risk factors have been proposed to play a role in the etiopathogenesis of schizophrenia. These include season of birth, maternal infections, obstetric complications, adverse events at early childhood, and drug abuse. Despite the progress in identification of genetic and environmental risk factors, we still have a limited understanding of the mechanisms whereby gene-environment interactions (G × E) operate in schizophrenia and psychoses at large. In this review we provide a critical analysis of current animal models of G × E relevant to psychotic disorders and propose that dimensional perspective will advance our understanding of the complex mechanisms of these disorders.
Collapse
Affiliation(s)
- Yavuz Ayhan
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, USA; Hacettepe University Faculty of Medicine, Department of Psychiatry, Turkey
| | - Ross McFarland
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, USA; Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, USA
| | - Mikhail V Pletnikov
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, USA; Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, USA; Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, USA; Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, USA.
| |
Collapse
|