101
|
DNA sequence-level analyses reveal potential phenotypic modifiers in a large family with psychiatric disorders. Mol Psychiatry 2018; 23:2254-2265. [PMID: 29880880 PMCID: PMC6294736 DOI: 10.1038/s41380-018-0087-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 03/30/2018] [Accepted: 04/09/2018] [Indexed: 02/07/2023]
Abstract
Psychiatric disorders are a group of genetically related diseases with highly polygenic architectures. Genome-wide association analyses have made substantial progress towards understanding the genetic architecture of these disorders. More recently, exome- and whole-genome sequencing of cases and families have identified rare, high penetrant variants that provide direct functional insight. There remains, however, a gap in the heritability explained by these complementary approaches. To understand how multiple genetic variants combine to modify both severity and penetrance of a highly penetrant variant, we sequenced 48 whole genomes from a family with a high loading of psychiatric disorder linked to a balanced chromosomal translocation. The (1;11)(q42;q14.3) translocation directly disrupts three genes: DISC1, DISC2, DISC1FP and has been linked to multiple brain imaging and neurocognitive outcomes in the family. Using DNA sequence-level linkage analysis, functional annotation and population-based association, we identified common and rare variants in GRM5 (minor allele frequency (MAF) > 0.05), PDE4D (MAF > 0.2) and CNTN5 (MAF < 0.01) that may help explain the individual differences in phenotypic expression in the family. We suggest that whole-genome sequencing in large families will improve the understanding of the combined effects of the rare and common sequence variation underlying psychiatric phenotypes.
Collapse
|
102
|
Chen X, Ku L, Mei R, Liu G, Xu C, Wen Z, Zhao X, Wang F, Xiao L, Feng Y. Novel schizophrenia risk factor pathways regulate FEZ1 to advance oligodendroglia development. Transl Psychiatry 2017; 7:1293. [PMID: 29249816 PMCID: PMC5802537 DOI: 10.1038/s41398-017-0028-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/15/2017] [Accepted: 08/24/2017] [Indexed: 01/08/2023] Open
Abstract
Neuropsychiatric disorders, represented by schizophrenia, affect not only neurons but also myelinating oligodendroglia (OL), both contribute to the complex etiology. Although numerous susceptibility genes for schizophrenia have been identified, their function has been primarily studied in neurons. Whether malfunction of risk genes underlies OL defects in schizophrenia pathogenesis remains poorly understood. In this study, we investigated the function and regulation of the well-recognized schizophrenia risk factor, Fasciculation and Elongation Protein Zeta-1 (FEZ1), in OL. We found that FEZ1 is expressed in oligodendroglia progenitor cells (OPCs) derived from rodent brains and human induced pluripotent stem cells (iPSCs) in culture and in myelinating oligodendrocytes in the brain. In addition, a vigorous upregulation of FEZ1 occurs during OPC differentiation and myelinogenesis, whereas knockdown of FEZ1 significantly attenuates the development of OL process arbors. We further showed that transcription of the Fez1 gene in OL cells is governed by a sophisticated functional interplay between histone acetylation-mediated chromatin modification and transcription factors that are dysregulated in schizophrenia. At the post-transcriptional level, the selective RNA-binding protein QKI, a glia-specific risk factor of schizophrenia, binds FEZ1 mRNA. Moreover, QKI deficiency results in a marked reduction of FEZ1 specifically in OL cells of the quakingviable (qkv) hypomyelination mutant mice. These observations have uncovered novel pathways that involve multifaceted genetic lesions and/or epigenetic dysregulations in schizophrenia, which converge on FEZ1 regulation and cause OL impairment in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Xianjun Chen
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing, 400038, China
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Li Ku
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ruyi Mei
- Institute of Developmental and Regenerative Biology, Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 310036, China
| | - Guanglu Liu
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Chongchong Xu
- Department of Psychiatry and Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Zhexing Wen
- Department of Psychiatry and Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Xiaofeng Zhao
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Institute of Developmental and Regenerative Biology, Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 310036, China
| | - Fei Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing, 400038, China
| | - Lan Xiao
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing, 400038, China.
| | - Yue Feng
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
103
|
DISC1 Regulates Neurogenesis via Modulating Kinetochore Attachment of Ndel1/Nde1 during Mitosis. Neuron 2017; 96:1041-1054.e5. [PMID: 29103808 DOI: 10.1016/j.neuron.2017.10.010] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 09/18/2017] [Accepted: 10/05/2017] [Indexed: 02/08/2023]
Abstract
Mutations of DISC1 (disrupted-in-schizophrenia 1) have been associated with major psychiatric disorders. Despite the hundreds of DISC1-binding proteins reported, almost nothing is known about how DISC1 interacts with other proteins structurally to impact human brain development. Here we solved the high-resolution structure of DISC1 C-terminal tail in complex with its binding domain of Ndel1. Mechanistically, DISC1 regulates Ndel1's kinetochore attachment, but not its centrosome localization, during mitosis. Functionally, disrupting DISC1/Ndel1 complex formation prolongs mitotic length and interferes with cell-cycle progression in human cells, and it causes cell-cycle deficits of radial glial cells in the embryonic mouse cortex and human forebrain organoids. We also observed similar deficits in organoids derived from schizophrenia patient induced pluripotent stem cells (iPSCs) with a DISC1 mutation that disrupts its interaction with Ndel1. Our study uncovers a new mechanism of action for DISC1 based on its structure, and it has implications for how genetic insults may contribute to psychiatric disorders.
Collapse
|
104
|
Intra-nasal dopamine alleviates cognitive deficits in tgDISC1 rats which overexpress the human DISC1 gene. Neurobiol Learn Mem 2017; 146:12-20. [PMID: 29107702 DOI: 10.1016/j.nlm.2017.10.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 10/26/2017] [Accepted: 10/27/2017] [Indexed: 01/15/2023]
Abstract
The Disrupted-in-Schizophrenia 1 (DISC1) gene has been associated with mental illnesses such as major depression and schizophrenia. The transgenic DISC1 (tgDISC1) rat, which overexpresses the human DISC1 gene, is known to exhibit deficient dopamine (DA) homeostasis. To ascertain whether the DISC1 gene also impacts cognitive functions, 14-15 months old male tgDISC1 rats and wild-type controls were subjected to the novel object preference (NOP) test and the object-based attention test (OBAT) in order to assess short-term memory (1 h), long-term memory (24 h), and attention. RESULTS The tgDISC1 group exhibited intact short-term memory, but deficient long-term-memory in the NOP test and deficient attention-related behavior in the OBAT. In a different group of tgDISC1 rats, 3 mg/kg intranasally applied dopamine (IN-DA) or its vehicle was applied prior to the NOP or the OBAT test. IN-DA reversed cognitive deficits in both the NOP and OBAT tests. In a further cohort of tgDISC1 rats, post-mortem levels of DA, noradrenaline, serotonin and acetylcholine were determined in a variety of brain regions. The tgDISC1 group had less DA in the neostriatum, hippocampus and amygdala, less acetylcholine in neostriatum, nucleus accumbens, hippocampus, and amygdala, more serotonin in the nucleus accumbens, and less serotonin and noradrenaline in the amygdala. CONCLUSIONS Our findings show that DISC1 overexpression and misassembly is associated with deficits in long-term memory and attention-related behavior. Since behavioral impairments in tgDISC1 rats were reversed by IN-DA, DA deficiency may be a major cause for the behavioral deficits expressed in this model.
Collapse
|
105
|
Eachus H, Bright C, Cunliffe VT, Placzek M, Wood JD, Watt PJ. Disrupted-in-Schizophrenia-1 is essential for normal hypothalamic-pituitary-interrenal (HPI) axis function. Hum Mol Genet 2017; 26:1992-2005. [PMID: 28334933 PMCID: PMC5437527 DOI: 10.1093/hmg/ddx076] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 02/23/2017] [Indexed: 02/01/2023] Open
Abstract
Psychiatric disorders arise due to an interplay of genetic and environmental factors, including stress. Studies in rodents have shown that mutants for Disrupted-In-Schizophrenia-1 (DISC1), a well-accepted genetic risk factor for mental illness, display abnormal behaviours in response to stress, but the mechanisms through which DISC1 affects stress responses remain poorly understood. Using two lines of zebrafish homozygous mutant for disc1, we investigated behaviour and functioning of the hypothalamic-pituitary-interrenal (HPI) axis, the fish equivalent of the hypothalamic-pituitary-adrenal (HPA) axis. Here, we show that the role of DISC1 in stress responses is evolutionarily conserved and that DISC1 is essential for normal functioning of the HPI axis. Adult zebrafish homozygous mutant for disc1 show aberrant behavioural responses to stress. Our studies reveal that in the embryo, disc1 is expressed in neural progenitor cells of the hypothalamus, a conserved region of the vertebrate brain that centrally controls responses to environmental stressors. In disc1 mutant embryos, proliferating rx3+ hypothalamic progenitors are not maintained normally and neuronal differentiation is compromised: rx3-derived ff1b+ neurons, implicated in anxiety-related behaviours, and corticotrophin releasing hormone (crh) neurons, key regulators of the stress axis, develop abnormally, and rx3-derived pomc+ neurons are disorganised. Abnormal hypothalamic development is associated with dysfunctional behavioural and neuroendocrine stress responses. In contrast to wild type siblings, disc1 mutant larvae show altered crh levels, fail to upregulate cortisol levels when under stress and do not modulate shoal cohesion, indicative of abnormal social behaviour. These data indicate that disc1 is essential for normal development of the hypothalamus and for the correct functioning of the HPA/HPI axis.
Collapse
Affiliation(s)
- Helen Eachus
- Department of Animal and Plant Sciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK.,The Bateson Centre, Department of Biomedical Science, Firth Court, Western Bank, Sheffield S10 2TN, UK
| | - Charlotte Bright
- Department of Animal and Plant Sciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Vincent T Cunliffe
- The Bateson Centre, Department of Biomedical Science, Firth Court, Western Bank, Sheffield S10 2TN, UK
| | - Marysia Placzek
- The Bateson Centre, Department of Biomedical Science, Firth Court, Western Bank, Sheffield S10 2TN, UK
| | - Jonathan D Wood
- The Bateson Centre, Department of Biomedical Science, Firth Court, Western Bank, Sheffield S10 2TN, UK.,Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Penelope J Watt
- Department of Animal and Plant Sciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| |
Collapse
|
106
|
Hartwig C, Monis WJ, Chen X, Dickman DK, Pazour GJ, Faundez V. Neurodevelopmental disease mechanisms, primary cilia, and endosomes converge on the BLOC-1 and BORC complexes. Dev Neurobiol 2017; 78:311-330. [PMID: 28986965 DOI: 10.1002/dneu.22542] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/08/2017] [Accepted: 09/15/2017] [Indexed: 12/12/2022]
Abstract
The biogenesis of lysosome-related organelles complex-1 (BLOC-1) and the bloc-one-related complex (BORC) are the cytosolic protein complexes required for specialized membrane protein traffic along the endocytic route and the spatial distribution of endosome-derived compartments, respectively. BLOC-1 and BORC complex subunits and components of their interactomes have been associated with the risk and/or pathomechanisms of neurodevelopmental disorders. Thus, cellular processes requiring BLOC-1 and BORC interactomes have the potential to offer novel insight into mechanisms underlying behavioral defects. We focus on interactions between BLOC-1 or BORC subunits with the actin and microtubule cytoskeleton, membrane tethers, and SNAREs. These interactions highlight requirements for BLOC-1 and BORC in membrane movement by motors, control of actin polymerization, and targeting of membrane proteins to specialized cellular domains such as the nerve terminal and the primary cilium. We propose that the endosome-primary cilia pathway is an underappreciated hub in the genesis and mechanisms of neurodevelopmental disorders. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 311-330, 2018.
Collapse
Affiliation(s)
- Cortnie Hartwig
- Department of Cell Biology, Emory University, Atlanta, Georgia, 30322
| | - William J Monis
- Program in Molecular Medicine, University of Massachusetts Medical School, Biotech II, Worcester, Massachusetts, 01605
| | - Xun Chen
- Department of Biology, Neurobiology Section, University of Southern California, Los Angeles, California, 90089
| | - Dion K Dickman
- Department of Biology, Neurobiology Section, University of Southern California, Los Angeles, California, 90089
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Biotech II, Worcester, Massachusetts, 01605
| | - Victor Faundez
- Department of Cell Biology, Emory University, Atlanta, Georgia, 30322
| |
Collapse
|
107
|
Terrillion CE, Abazyan B, Yang Z, Crawford J, Shevelkin AV, Jouroukhin Y, Yoo KH, Cho CH, Roychaudhuri R, Snyder SH, Jang MH, Pletnikov MV. DISC1 in Astrocytes Influences Adult Neurogenesis and Hippocampus-Dependent Behaviors in Mice. Neuropsychopharmacology 2017; 42. [PMID: 28631721 PMCID: PMC5603806 DOI: 10.1038/npp.2017.129] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The functional role of genetic variants in glia in the pathogenesis of psychiatric disorders remains poorly studied. Disrupted-In-Schizophrenia 1 (DISC1), a genetic risk factor implicated in major mental disorders, has been implicated in regulation of astrocyte functions. As both astrocytes and DISC1 influence adult neurogenesis in the dentate gyrus (DG) of the hippocampus, we hypothesized that selective expression of dominant-negative C-terminus-truncated human DISC1 (mutant DISC1) in astrocytes would affect adult hippocampal neurogenesis and hippocampus-dependent behaviors. A series of behavioral tests were performed in mice with or without expression of mutant DISC1 in astrocytes during late postnatal development. In conjunction with behavioral tests, we evaluated adult neurogenesis, including neural progenitor proliferation and dendrite development of newborn neurons in the DG. The ameliorative effects of D-serine on mutant DISC1-associated behaviors and abnormal adult neurogenesis were also examined. Expression of mutant DISC1 in astrocytes decreased neural progenitor proliferation and dendrite growth of newborn neurons, and produced elevated anxiety, attenuated social behaviors, and impaired hippocampus-dependent learning and memory. Chronic treatment with D-serine ameliorated the behavioral alterations and rescued abnormal adult neurogenesis in mutant DISC1 mice. Our findings suggest that psychiatric genetic risk factors expressed in astrocytes could affect adult hippocampal neurogenesis and contribute to aspects of psychiatric disease through abnormal production of D-serine.
Collapse
Affiliation(s)
- Chantelle E Terrillion
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bagrat Abazyan
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhongxi Yang
- Department of Neurologic Surgery, Mayo Clinic College of Medicine, Rochester, MN, USA,Department of Neurosurgery, First Hospital of Jilin University, Changchun, China
| | - Joshua Crawford
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexey V Shevelkin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yan Jouroukhin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ki Hyun Yoo
- Department of Neurologic Surgery, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Chang Hoon Cho
- Department of Neurologic Surgery, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Robin Roychaudhuri
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Solomon H Snyder
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mi-Hyeon Jang
- Department of Neurologic Surgery, Mayo Clinic College of Medicine, Rochester, MN, USA,Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Mikhail V Pletnikov
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA,Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA,Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA,Johns Hopkins University School of Medicine, 600 North Wolfe Street, CMSC 8-121, Baltimore, MD 21287, USA, Tel: +410-502-3760, Fax: +410-614-0013, E-mail:
| |
Collapse
|
108
|
Deng D, Jian C, Lei L, Zhou Y, McSweeney C, Dong F, Shen Y, Zou D, Wang Y, Wu Y, Zhang L, Mao Y. A prenatal interruption of DISC1 function in the brain exhibits a lasting impact on adult behaviors, brain metabolism, and interneuron development. Oncotarget 2017; 8:84798-84817. [PMID: 29156684 PMCID: PMC5689574 DOI: 10.18632/oncotarget.21381] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 09/03/2017] [Indexed: 02/03/2023] Open
Abstract
Mental illnesses like schizophrenia (SCZ) and major depression disorder (MDD) are devastating brain disorders. The SCZ risk gene, disrupted in schizophrenia 1 (DISC1), has been associated with neuropsychiatric conditions. However, little is known regarding the long-lasting impacts on brain metabolism and behavioral outcomes from genetic insults on fetal NPCs during early life. We have established a new mouse model that specifically interrupts DISC1 functions in NPCs in vivo by a dominant-negative DISC1 (DN-DISC1) with a precise temporal and spatial regulation. Interestingly, prenatal interruption of mouse Disc1 function in NPCs leads to abnormal depression-like deficit in adult mice. Here we took a novel unbiased metabonomics approach to identify brain-specific metabolites that are significantly changed in DN-DISC1 mice. Surprisingly, the inhibitory neurotransmitter, GABA, is augmented. Consistently, parvalbumin (PV) interneurons are increased in the cingulate cortex, retrosplenial granular cortex, and motor cortex. Interestingly, somatostatin (SST) positive and neuropeptide Y (NPY) interneurons are decreased in some brain regions, suggesting that DN-DISC1 expression affects the localization of interneuron subtypes. To further explore the cellular mechanisms that cause this change, DN-DISC1 suppresses proliferation and promotes the cell cycle exit of progenitors in the medial ganglionic eminence (MGE), whereas it stimulates ectopic proliferation of neighboring cells through cell non-autonomous effect. Mechanistically, it modulates GSK3 activity and interrupts Dlx2 activity in the Wnt activation. In sum, our results provide evidence that specific genetic insults on NSCs at a short period of time could lead to prolonged changes of brain metabolism and development, eventually behavioral defects.
Collapse
Affiliation(s)
- Dazhi Deng
- Department of Emergency, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China.,Department of Biology, Pennsylvania State University, University Park, PA, USA
| | - Chongdong Jian
- Department of Biology, Pennsylvania State University, University Park, PA, USA.,Department of Neurology, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Ling Lei
- Department of Biology, Pennsylvania State University, University Park, PA, USA.,Health Examination Center, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Yijing Zhou
- Department of Biology, Pennsylvania State University, University Park, PA, USA
| | - Colleen McSweeney
- Department of Biology, Pennsylvania State University, University Park, PA, USA
| | - Fengping Dong
- Department of Biology, Pennsylvania State University, University Park, PA, USA
| | - Yilun Shen
- Department of Biology, Pennsylvania State University, University Park, PA, USA
| | - Donghua Zou
- Department of Neurology, The First People's Hospital of Nanning, Nanning, Guangxi, China
| | - Yonggang Wang
- Department of Neurology, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yuan Wu
- Department of Neurology, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Limin Zhang
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Yingwei Mao
- Department of Biology, Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
109
|
Murphy LC, Millar JK. Regulation of mitochondrial dynamics by DISC1, a putative risk factor for major mental illness. Schizophr Res 2017; 187:55-61. [PMID: 28082141 DOI: 10.1016/j.schres.2016.12.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 12/22/2022]
Abstract
Mitochondria are dynamic organelles that are essential to power the process of neurotransmission. Neurons must therefore ensure that mitochondria maintain their functional integrity and are efficiently transported along the full extent of the axons and dendrites, from soma to synapses. Mitochondrial dynamics (trafficking, fission and fusion) co-ordinately regulate mitochondrial quality control and function. DISC1 is a component of the mitochondrial transport machinery and regulates mitochondrial dynamics. DISC1's role in this is adversely affected by sequence variants connected to brain structure/function and disease risk, and by mutant truncation. The DISC1 interactors NDE1 and GSK3β are also involved, indicating a convergence of putative risk factors for psychiatric illness upon mitochondrial dynamics.
Collapse
Affiliation(s)
- Laura C Murphy
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetic and Molecular Medicine at the University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - J Kirsty Millar
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetic and Molecular Medicine at the University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK.
| |
Collapse
|
110
|
Liu YN, Lu SY, Yao J. Application of induced pluripotent stem cells to understand neurobiological basis of bipolar disorder and schizophrenia. Psychiatry Clin Neurosci 2017; 71:579-599. [PMID: 28393474 DOI: 10.1111/pcn.12528] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/04/2017] [Indexed: 12/12/2022]
Abstract
The etiology of neuropsychiatric disorders, such as schizophrenia and bipolar disorder, usually involves complex combinations of genetic defects/variations and environmental impacts, which hindered, for a long time, research efforts based on animal models and patients' non-neuronal cells or post-mortem tissues. However, the development of human induced pluripotent stem cell (iPSC) technology by the Yamanaka group was immediately applied to establish cell research models for neuronal disorders. Since then, techniques to achieve highly efficient differentiation of different types of neural cells following iPSC modeling have made much progress. The fast-growing iPSC and neural differentiation techniques have brought valuable insights into the pathology and neurobiology of neuropsychiatric disorders. In this article, we first review the application of iPSC technology in modeling neuronal disorders and discuss the progress in the accompanying neural differentiation. Then, we summarize the progress in iPSC-based research that has been accomplished so far regarding schizophrenia and bipolar disorder.
Collapse
Affiliation(s)
- Yao-Nan Liu
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Si-Yao Lu
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Jun Yao
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| |
Collapse
|
111
|
Wu Q, Tang W, Luo Z, Li Y, Shu Y, Yue Z, Xiao B, Feng L. DISC1 Regulates the Proliferation and Migration of Mouse Neural Stem/Progenitor Cells through Pax5, Sox2, Dll1 and Neurog2. Front Cell Neurosci 2017; 11:261. [PMID: 28900388 PMCID: PMC5581844 DOI: 10.3389/fncel.2017.00261] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/11/2017] [Indexed: 11/27/2022] Open
Abstract
Background: Disrupted-in-schizophrenia 1 (DISC1) regulates neurogenesis and is a genetic risk factor for major psychiatric disorders. However, how DISC1 dysfunction affects neurogenesis and cell cycle progression at the molecular level is still unknown. Here, we investigated the role of DISC1 in regulating proliferation, migration, cell cycle progression and apoptosis in mouse neural stem/progenitor cells (MNSPCs) in vitro. Methods: MNSPCs were isolated and cultured from mouse fetal hippocampi. Retroviral vectors or siRNAs were used to manipulate DISC1 expression in MNSPCs. Proliferation, migration, cell cycle progression and apoptosis of altered MNSPCs were analyzed in cell proliferation assays (MTS), transwell system and flow cytometry. A neurogenesis specific polymerase chain reaction (PCR) array was used to identify genes downstream of DISC1, and functional analysis was performed through transfection of expression plasmids and siRNAs. Results: Loss of DISC1 reduced proliferation and migration of MNSPCs, while an increase in DISC1 led to increased proliferation and migration. Meanwhile, an increase in the proportion of cells in G0/G1 phase was concomitant with reduced levels of DISC1, but significant changes were not observed in the number MNSPCs undergoing apoptosis. Paired box gene 5 (Pax5), sex determining region Y-box 2 (Sox2), delta-like1 (Dll1) and Neurogenin2 (Neurog2) emerged as candidate molecules downstream of DISC1, and rescue experiments demonstrated that increased or decreased expression of either molecule regulated proliferation and migration in DISC1-altered MNSPCs. Conclusion: These results suggest that Pax5, Sox2, Dll1 and Neurog2 mediate DISC1 activity in MNSPC proliferation and migration.
Collapse
Affiliation(s)
- Qian Wu
- Department of Neurology, First Affiliated Hospital, Kunming Medical UniversityKunming, China
- Department of Neurology, Xiangya Hospital, Central South UniversityChangsha, China
| | - Weiting Tang
- Department of Neurology, Xiangya Hospital, Central South UniversityChangsha, China
| | - Zhaohui Luo
- Department of Neurology, Xiangya Hospital, Central South UniversityChangsha, China
| | - Yi Li
- Department of Neurology, University of Massachusetts Medical SchoolWorcester, MA, United States
| | - Yi Shu
- Department of Neurology, The Second Xiangya Hospital, Central South UniversityChangsha, China
| | - Zongwei Yue
- Department of Neurology, Xiangya Hospital, Central South UniversityChangsha, China
- Department of Neurology, Yale University School of MedicineNew Haven, CT, United States
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South UniversityChangsha, China
| | - Li Feng
- Department of Neurology, Xiangya Hospital, Central South UniversityChangsha, China
- Department of Neurology, Yale University School of MedicineNew Haven, CT, United States
| |
Collapse
|
112
|
Smith AK, Jovanovic T, Kilaru V, Lori A, Gensler L, Lee SS, Norrholm SD, Massa N, Cuthbert B, Bradley B, Ressler KJ, Duncan E. A Gene-Based Analysis of Acoustic Startle Latency. Front Psychiatry 2017; 8:117. [PMID: 28729842 PMCID: PMC5498475 DOI: 10.3389/fpsyt.2017.00117] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 06/19/2017] [Indexed: 12/16/2022] Open
Abstract
Latency of the acoustic startle response is the time required from the presentation of startling auditory stimulus until the startle response is elicited and provides an index of neural processing speed. Latency is prolonged in subjects with schizophrenia compared to controls in some but not all studies and is 68-90% heritable in baseline startle trials. In order to determine the genetic association with latency as a potential inroad into genetically based vulnerability to psychosis, we conducted a gene-based study of latency followed by an independent replication study of significant gene findings with a single-nucleotide polymorphism (SNP)-based analysis of schizophrenia and control subjects. 313 subjects from an urban population of low socioeconomic status with mixed psychiatric diagnoses were included in the gene-based study. Startle testing was conducted using a Biopac M150 system according to our published methods. Genotyping was performed with the Omni-Quad 1M or the Omni Express BeadChip. The replication study was conducted on 154 schizophrenia subjects and 123 psychiatric controls. Genetic analyses were conducted with Illumina Human Omni1-Quad and OmniExpress BeadChips. Twenty-nine SNPs were selected from four genes that were significant in the gene-based analysis and also associated with startle and/or schizophrenia in the literature. Linear regressions on latency were conducted, controlling for age, race, and diagnosis as a dichotomous variable. In the gene-based study, 2,870 genes demonstrated the evidence of association after correction for multiple comparisons (false discovery rate < 0.05). Pathway analysis of these genes revealed enrichment for relevant biological processes including neural transmission (p = 0.0029), synaptic transmission (p = 0.0032), and neuronal development (p = 0.024). The subsequent SNP-based replication analysis revealed a strong association of onset latency with the SNP rs901561 on the neuregulin gene (NRG1) in an additive model (beta = 0.21, p = 0.001), indicating that subjects with the AA and AG genotypes had slower mean latency than subjects with GG genotype. In conclusion, startle latency, a highly heritable measure that is slowed in schizophrenia, may be a useful biological probe for genetic contributions to psychotic disorders. Our analyses in two independent populations point to a significant prediction of startle latency by genetic variation in NRG1.
Collapse
Affiliation(s)
- Alicia K. Smith
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, United States
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States
| | - Tanja Jovanovic
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States
| | - Varun Kilaru
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States
| | - Adriana Lori
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States
| | - Lauren Gensler
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States
| | - Samuel S. Lee
- Department of Emergency Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Seth Davin Norrholm
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States
- Mental Health Service Line, Atlanta Veterans Affairs Medical Center, Decatur, GA, United States
| | - Nicholas Massa
- Mental Health Service Line, Atlanta Veterans Affairs Medical Center, Decatur, GA, United States
| | - Bruce Cuthbert
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States
- Mental Health Service Line, Atlanta Veterans Affairs Medical Center, Decatur, GA, United States
| | - Bekh Bradley
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States
- Mental Health Service Line, Atlanta Veterans Affairs Medical Center, Decatur, GA, United States
| | - Kerry J. Ressler
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
| | - Erica Duncan
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States
- Mental Health Service Line, Atlanta Veterans Affairs Medical Center, Decatur, GA, United States
| |
Collapse
|
113
|
Abstract
Human genetic studies have been the driving force in bringing to light the underlying biology of psychiatric conditions. As these studies fill in the gaps in our knowledge of the mechanisms at play, we will be better equipped to design therapies in rational and targeted ways, or repurpose existing therapies in previously unanticipated ways. This review is intended for those unfamiliar with psychiatric genetics as a field and provides a primer on different modes of genetic variation, the technologies currently used to probe them, and concepts that provide context for interpreting the gene-phenotype relationship. Like other subfields in human genetics, psychiatric genetics is moving from microarray technology to sequencing-based approaches as barriers of cost and expertise are removed, and the ramifications of this transition are discussed here. A summary is then given of recent genetic discoveries in a number of neuropsychiatric conditions, with particular emphasis on neurodevelopmental conditions. The general impact of genetics on drug development has been to underscore the extensive etiological heterogeneity in seemingly cohesive diagnostic categories. Consequently, the path forward is not in therapies hoping to reach large swaths of patients sharing a clinically defined diagnosis, but rather in targeting patients belonging to specific "biotypes" defined through a combination of objective, quantifiable data, including genotype.
Collapse
Affiliation(s)
- Jacob J Michaelson
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
- Department of Biomedical Engineering, University of Iowa College of Engineering, Iowa City, IA, USA.
- Department of Communication Sciences and Disorders, University of Iowa College of Liberal Arts and Sciences, Iowa City, IA, USA.
- Iowa Institute of Human Genetics, University of Iowa, Iowa City, IA, USA.
- Genetics Cluster Initiative, University of Iowa, Iowa City, IA, USA.
- The DeLTA Center, University of Iowa, Iowa City, IA, USA.
- University of Iowa Informatics Initiative, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
114
|
Hoekstra SD, Stringer S, Heine VM, Posthuma D. Genetically-Informed Patient Selection for iPSC Studies of Complex Diseases May Aid in Reducing Cellular Heterogeneity. Front Cell Neurosci 2017; 11:164. [PMID: 28659762 PMCID: PMC5468546 DOI: 10.3389/fncel.2017.00164] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 05/29/2017] [Indexed: 01/27/2023] Open
Abstract
Induced pluripotent stem cell (iPSC) technology is more and more used for the study of genetically complex human disease but is challenged by variability, sample size and polygenicity. We discuss studies involving iPSC-derived neurons from patients with Schizophrenia (SCZ), to exemplify that heterogeneity in sampling strategy complicate the detection of disease mechanisms. We offer a solution to controlling variability within and between iPSC studies by using specific patient selection strategies.
Collapse
Affiliation(s)
- Stephanie D Hoekstra
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit AmsterdamAmsterdam, Netherlands
| | - Sven Stringer
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit AmsterdamAmsterdam, Netherlands
| | - Vivi M Heine
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit AmsterdamAmsterdam, Netherlands.,Department Pediatrics/Child Neurology, VU University Medical CenterAmsterdam, Netherlands
| | - Danielle Posthuma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit AmsterdamAmsterdam, Netherlands.,Department of Clinical Genetics, VU University Medical Center, Amsterdam NeuroscienceAmsterdam, Netherlands
| |
Collapse
|
115
|
Expression of mutant DISC1 in Purkinje cells increases their spontaneous activity and impairs cognitive and social behaviors in mice. Neurobiol Dis 2017; 103:144-153. [PMID: 28392471 DOI: 10.1016/j.nbd.2017.04.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 04/02/2017] [Accepted: 04/05/2017] [Indexed: 12/15/2022] Open
Abstract
In addition to motor function, the cerebellum has been implicated in cognitive and social behaviors. Various structural and functional abnormalities of Purkinje cells (PCs) have been observed in schizophrenia and autism. As PCs express the gene Disrupted-In-Schizophrenia-1 (DISC1), and DISC1 variants have been associated with neurodevelopmental disorders, we evaluated the role of DISC1 in cerebellar physiology and associated behaviors using a mouse model of inducible and selective expression of a dominant-negative, C-terminus truncated human DISC1 (mutant DISC1) in PCs. Mutant DISC1 male mice demonstrated impaired social and novel placement recognition. No group differences were found in novelty-induced hyperactivity, elevated plus maze test, spontaneous alternation, spatial recognition in Y maze, sociability or accelerated rotarod. Expression of mutant DISC1 was associated with a decreased number of large somata PCs (volume: 3000-5000μm3) and an increased number of smaller somata PCs (volume: 750-1000μm3) without affecting the total number of PCs or the volume of the cerebellum. Compared to control mice, attached loose patch recordings of PCs in mutant DISC1 mice revealed increased spontaneous firing of PCs; and whole cell recordings showed increased amplitude and frequency of mEPSCs without significant changes in either Rinput or parallel fiber EPSC paired-pulse ratio. Our findings indicate that mutant DISC1 alters the physiology of PCs, possibly leading to abnormal recognition memory in mice.
Collapse
|
116
|
Bradshaw NJ, Hayashi MAF. NDE1 and NDEL1 from genes to (mal)functions: parallel but distinct roles impacting on neurodevelopmental disorders and psychiatric illness. Cell Mol Life Sci 2017; 74:1191-1210. [PMID: 27742926 PMCID: PMC11107680 DOI: 10.1007/s00018-016-2395-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 09/13/2016] [Accepted: 10/06/2016] [Indexed: 01/01/2023]
Abstract
NDE1 (Nuclear Distribution Element 1, also known as NudE) and NDEL1 (NDE-Like 1, also known as NudEL) are the mammalian homologues of the fungus nudE gene, with important and at least partially overlapping roles for brain development. While a large number of studies describe the various properties and functions of these proteins, many do not directly compare the similarities and differences between NDE1 and NDEL1. Although sharing a high degree structural similarity and multiple common cellular roles, each protein presents several distinct features that justify their parallel but also unique functions. Notably both proteins have key binding partners in dynein, LIS1 and DISC1, which impact on neurodevelopmental and psychiatric illnesses. Both are implicated in schizophrenia through genetic and functional evidence, with NDE1 also strongly implicated in microcephaly, as well as other neurodevelopmental and psychiatric conditions through copy number variation, while NDEL1 possesses an oligopeptidase activity with a unique potential as a biomarker in schizophrenia. In this review, we aim to give a comprehensive overview of the various cellular roles of these proteins in a "bottom-up" manner, from their biochemistry and protein-protein interactions on the molecular level, up to the consequences for neuronal differentiation, and ultimately to their importance for correct cortical development, with direct consequences for the pathophysiology of neurodevelopmental and mental illness.
Collapse
Affiliation(s)
- Nicholas J Bradshaw
- Department of Neuropathology, Heinrich Heine University, Düsseldorf, Germany.
| | - Mirian A F Hayashi
- Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, Brazil
| |
Collapse
|
117
|
Morosawa S, Iritani S, Fujishiro H, Sekiguchi H, Torii Y, Habuchi C, Kuroda K, Kaibuchi K, Ozaki N. Neuropeptide Y neuronal network dysfunction in the frontal lobe of a genetic mouse model of schizophrenia. Neuropeptides 2017; 62:27-35. [PMID: 28073591 DOI: 10.1016/j.npep.2016.12.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/18/2016] [Accepted: 12/22/2016] [Indexed: 11/23/2022]
Abstract
Neuropeptide Y (NPY) has been found to play a critical role in various mental functions as a neurotransmitter and is involved in the development of schizophrenia, a particularly intractable psychiatric disease whose precise etiology remains unknown. Recent molecular biological investigations have identified several candidate genes which may be associated with this disease, including disrupted-in-schizophrenia 1 (DISC1). The role of DISC1 would involve neurogenesis and neuronal migration. However, the functional consequences of this gene defect have not yet been fully clarified in neuronal systems. In the present study, to clarify the neuropathological changes associated with the function of DISC1, we explored how DISC1 dysfunction can induce abnormalities in the NPY neuronal network in the central nervous system. We performed immunohistochemical analyses (including the observation of the distribution and density) of prefrontal cortex specimens from DISC1-knockout (KO) mice, which are considered to be a novel animal model of schizophrenia. We then evaluated the number and size of NPY-immunoreactive (NPY-IR) neurons and the length of NPY-IR fibers. The number of NPY-IR neurons and the length of the fibers were decreased in the prefrontal cortex of DISC1-KO mice. The decrease was particularly prominent in the superficial regions, and the distribution of NPY-IR neurons differed between wild-type and DISC1-KO mice. However, the size of the neurons in the cortices of the DISC1-KO and wild-type mice did not differ markedly. Our findings suggest that dysfunction of DISC1 may lead to the alteration of NPY neurons and neurotransmission issues in NPY-containing neuron systems, which seem to play important roles in both the mental function and neuronal development. DISC1 dysfunction may be involved in the pathogenesis of schizophrenia through the impairment of the NPY neuronal network.
Collapse
Affiliation(s)
- Shunsuke Morosawa
- Department of Psychiatry, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya, Aichi 466-8550, Japan.
| | - Shuji Iritani
- Department of Psychiatry, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya, Aichi 466-8550, Japan.
| | - Hiroshige Fujishiro
- Department of Psychiatry, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya, Aichi 466-8550, Japan.
| | - Hirotaka Sekiguchi
- Department of Psychiatry, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya, Aichi 466-8550, Japan.
| | - Youta Torii
- Department of Psychiatry, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya, Aichi 466-8550, Japan.
| | - Chikako Habuchi
- Department of Psychiatry, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya, Aichi 466-8550, Japan.
| | - Keisuke Kuroda
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya, Aichi 466-8550, Japan.
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya, Aichi 466-8550, Japan.
| | - Norio Ozaki
- Department of Psychiatry, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya, Aichi 466-8550, Japan.
| |
Collapse
|
118
|
Kitanishi T, Ito HT, Hayashi Y, Shinohara Y, Mizuseki K, Hikida T. Network mechanisms of hippocampal laterality, place coding, and goal-directed navigation. J Physiol Sci 2017; 67:247-258. [PMID: 27864684 PMCID: PMC10717435 DOI: 10.1007/s12576-016-0502-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 11/07/2016] [Indexed: 12/23/2022]
Abstract
The hippocampus and associated structures are responsible for episodic memory in humans. In rodents, the most prominent behavioral correlate of hippocampal neural activity is place coding, which is thought to underlie spatial navigation. While episodic memory is considered to be unique to humans in a restricted context, it has been proposed that the same neural circuitry and algorithms that enable spatial coding and navigation also support episodic memory. Here we review the recent progress in neural circuit mechanisms of hippocampal activity by introducing several topics: (1) cooperation and specialization of the bilateral hippocampi, (2) the role of synaptic plasticity in gamma phase-locking of spikes and place cell formation, (3) impaired goal-related activity and oscillations in a mouse model of mental disorders, and (4) a prefrontal-thalamo-hippocampal circuit for goal-directed spatial navigation.
Collapse
Affiliation(s)
- Takuma Kitanishi
- Department of Physiology, Osaka City University Graduate School of Medicine, Asahimachi 1-4-3, Abeno-ku, Osaka, 545-8585, Japan
- Center for Brain Science, Osaka City University Graduate School of Medicine, Asahimachi 1-4-3, Abeno-ku, Osaka, 545-8585, Japan
| | - Hiroshi T Ito
- Max Planck Institute for Brain Research, 60438, Frankfurt am Main, Germany
| | - Yuichiro Hayashi
- Frontier Research Center for Post-genome Science and Technology, Hokkaido University, Hokkaido, 001-0021, Japan
| | - Yoshiaki Shinohara
- Laboratory for Neuron-Glia Circuitry, RIKEN Brain Science Institute, Saitama, 351-0198, Japan
| | - Kenji Mizuseki
- Department of Physiology, Osaka City University Graduate School of Medicine, Asahimachi 1-4-3, Abeno-ku, Osaka, 545-8585, Japan.
- Center for Brain Science, Osaka City University Graduate School of Medicine, Asahimachi 1-4-3, Abeno-ku, Osaka, 545-8585, Japan.
| | - Takatoshi Hikida
- Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
119
|
Emerging Synaptic Molecules as Candidates in the Etiology of Neurological Disorders. Neural Plast 2017; 2017:8081758. [PMID: 28331639 PMCID: PMC5346360 DOI: 10.1155/2017/8081758] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 02/06/2017] [Indexed: 01/06/2023] Open
Abstract
Synapses are complex structures that allow communication between neurons in the central nervous system. Studies conducted in vertebrate and invertebrate models have contributed to the knowledge of the function of synaptic proteins. The functional synapse requires numerous protein complexes with specialized functions that are regulated in space and time to allow synaptic plasticity. However, their interplay during neuronal development, learning, and memory is poorly understood. Accumulating evidence links synapse proteins to neurodevelopmental, neuropsychiatric, and neurodegenerative diseases. In this review, we describe the way in which several proteins that participate in cell adhesion, scaffolding, exocytosis, and neurotransmitter reception from presynaptic and postsynaptic compartments, mainly from excitatory synapses, have been associated with several synaptopathies, and we relate their functions to the disease phenotype.
Collapse
|
120
|
Zai G, Robbins TW, Sahakian BJ, Kennedy JL. A review of molecular genetic studies of neurocognitive deficits in schizophrenia. Neurosci Biobehav Rev 2017; 72:50-67. [DOI: 10.1016/j.neubiorev.2016.10.024] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 10/17/2016] [Accepted: 10/27/2016] [Indexed: 02/08/2023]
|
121
|
Chen CY, Liu HY, Hsueh YP. TLR3 downregulates expression of schizophrenia gene Disc1 via MYD88 to control neuronal morphology. EMBO Rep 2016; 18:169-183. [PMID: 27979975 PMCID: PMC5210159 DOI: 10.15252/embr.201642586] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 11/08/2016] [Accepted: 11/09/2016] [Indexed: 01/28/2023] Open
Abstract
Viral infection during fetal or neonatal stages increases the risk of developing neuropsychiatric disorders such as schizophrenia and autism spectrum disorders. Although neurons express several key regulators of innate immunity, the role of neuronal innate immunity in psychiatric disorders is still unclear. Using cultured neurons and in vivo mouse brain studies, we show here that Toll‐like receptor 3 (TLR3) acts through myeloid differentiation primary response gene 88 (MYD88) to negatively control Disrupted in schizophrenia 1 (Disc1) expression, resulting in impairment of neuronal development. Cytokines are not involved in TLR3‐mediated inhibition of dendrite outgrowth. Instead, TLR3 signaling suppresses expression of several psychiatric disorder‐related genes, including Disc1. The impaired dendritic arborization caused by TLR3 activation is rescued by MYD88 deficiency or DISC1 overexpression. In addition, TLR3 activation at the neonatal stage increases dendritic spine density, but narrows spine heads at postnatal day 21 (P21), suggesting a long‐lasting effect of TLR3 activation on spinogenesis. Our study reveals a novel mechanism of TLR3 in regulation of dendritic morphology and provides an explanation for how environmental factors influence mental health.
Collapse
Affiliation(s)
- Chiung-Ya Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Hsin-Yu Liu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Yi-Ping Hsueh
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
122
|
Altered cytokine profile, pain sensitivity, and stress responsivity in mice with co-disruption of the developmental genes Neuregulin-1×DISC1. Behav Brain Res 2016; 320:113-118. [PMID: 27916686 DOI: 10.1016/j.bbr.2016.11.049] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 11/15/2016] [Accepted: 11/28/2016] [Indexed: 12/14/2022]
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, IL1β 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 IL1β. 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.
Collapse
|
123
|
Misassembly of full-length Disrupted-in-Schizophrenia 1 protein is linked to altered dopamine homeostasis and behavioral deficits. Mol Psychiatry 2016; 21:1561-1572. [PMID: 26754951 PMCID: PMC5078859 DOI: 10.1038/mp.2015.194] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 10/16/2015] [Accepted: 10/22/2015] [Indexed: 12/21/2022]
Abstract
Disrupted-in-schizophrenia 1 (DISC1) is a mental illness gene first identified in a Scottish pedigree. So far, DISC1-dependent phenotypes in animal models have been confined to expressing mutant DISC1. Here we investigated how pathology of full-length DISC1 protein could be a major mechanism in sporadic mental illness. We demonstrate that a novel transgenic rat model, modestly overexpressing the full-length DISC1 transgene, showed phenotypes consistent with a significant role of DISC1 misassembly in mental illness. The tgDISC1 rat displayed mainly perinuclear DISC1 aggregates in neurons. Furthermore, the tgDISC1 rat showed a robust signature of behavioral phenotypes that includes amphetamine supersensitivity, hyperexploratory behavior and rotarod deficits, all pointing to changes in dopamine (DA) neurotransmission. To understand the etiology of the behavioral deficits, we undertook a series of molecular studies in the dorsal striatum of tgDISC1 rats. We observed an 80% increase in high-affinity DA D2 receptors, an increased translocation of the dopamine transporter to the plasma membrane and a corresponding increase in DA inflow as observed by cyclic voltammetry. A reciprocal relationship between DISC1 protein assembly and DA homeostasis was corroborated by in vitro studies. Elevated cytosolic dopamine caused an increase in DISC1 multimerization, insolubility and complexing with the dopamine transporter, suggesting a physiological mechanism linking DISC1 assembly and dopamine homeostasis. DISC1 protein pathology and its interaction with dopamine homeostasis is a novel cellular mechanism that is relevant for behavioral control and may have a role in mental illness.
Collapse
|
124
|
Neuregulin-1 Regulates Cortical Inhibitory Neuron Dendrite and Synapse Growth through DISC1. Neural Plast 2016; 2016:7694385. [PMID: 27847649 PMCID: PMC5099462 DOI: 10.1155/2016/7694385] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 07/05/2016] [Accepted: 09/05/2016] [Indexed: 11/20/2022] Open
Abstract
Cortical inhibitory neurons play crucial roles in regulating excitatory synaptic networks and cognitive function and aberrant development of these cells have been linked to neurodevelopmental disorders. The secreted neurotrophic factor Neuregulin-1 (NRG1) and its receptor ErbB4 are established regulators of inhibitory neuron connectivity, but the developmental signalling mechanisms regulating this process remain poorly understood. Here, we provide evidence that NRG1-ErbB4 signalling functions through the multifunctional scaffold protein, Disrupted in Schizophrenia 1 (DISC1), to regulate the development of cortical inhibitory interneuron dendrite and synaptic growth. We found that NRG1 increases inhibitory neuron dendrite complexity and glutamatergic synapse formation onto inhibitory neurons and that this effect is blocked by expression of a dominant negative DISC1 mutant, or DISC1 knockdown. We also discovered that NRG1 treatment increases DISC1 expression and its localization to glutamatergic synapses being made onto cortical inhibitory neurons. Mechanistically, we determined that DISC1 binds ErbB4 within cortical inhibitory neurons. Collectively, these data suggest that a NRG1-ErbB4-DISC1 signalling pathway regulates the development of cortical inhibitory neuron dendrite and synaptic growth. Given that NRG1, ErbB4, and DISC1 are schizophrenia-linked genes, these findings shed light on how independent risk factors may signal in a common developmental pathway that contributes to neural connectivity defects and disease pathogenesis.
Collapse
|
125
|
Simultaneous effects on parvalbumin-positive interneuron and dopaminergic system development in a transgenic rat model for sporadic schizophrenia. Sci Rep 2016; 6:34946. [PMID: 27721451 PMCID: PMC5056355 DOI: 10.1038/srep34946] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/20/2016] [Indexed: 11/08/2022] Open
Abstract
To date, unequivocal neuroanatomical features have been demonstrated neither for sporadic nor for familial schizophrenia. Here, we investigated the neuroanatomical changes in a transgenic rat model for a subset of sporadic chronic mental illness (CMI), which modestly overexpresses human full-length, non-mutant Disrupted-in-Schizophrenia 1 (DISC1), and for which aberrant dopamine homeostasis consistent with some schizophrenia phenotypes has previously been reported. Neuroanatomical analysis revealed a reduced density of dopaminergic neurons in the substantia nigra and reduced dopaminergic fibres in the striatum. Parvalbumin-positive interneuron occurrence in the somatosensory cortex was shifted from layers II/III to V/VI, and the number of calbindin-positive interneurons was slightly decreased. Reduced corpus callosum thickness confirmed trend-level observations from in vivo MRI and voxel-wise tensor based morphometry. These neuroanatomical changes help explain functional phenotypes of this animal model, some of which resemble changes observed in human schizophrenia post mortem brain tissues. Our findings also demonstrate how a single molecular factor, DISC1 overexpression or misassembly, can account for a variety of seemingly unrelated morphological phenotypes and thus provides a possible unifying explanation for similar findings observed in sporadic schizophrenia patients. Our anatomical investigation of a defined model for sporadic mental illness enables a clearer definition of neuroanatomical changes associated with subsets of human sporadic schizophrenia.
Collapse
|
126
|
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: 3.1] [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
|
127
|
Phillips WA, Larkum ME, Harley CW, Silverstein SM. The effects of arousal on apical amplification and conscious state. Neurosci Conscious 2016; 2016:niw015. [PMID: 29877512 PMCID: PMC5934888 DOI: 10.1093/nc/niw015] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/28/2016] [Accepted: 08/08/2016] [Indexed: 01/19/2023] Open
Abstract
Neocortical pyramidal cells can integrate two classes of input separately and use one to modulate response to the other. Their tuft dendrites are electrotonically separated from basal dendrites and soma by the apical dendrite, and apical hyperpolarization-activated currents (Ih) further isolate subthreshold integration of tuft inputs. When apical depolarization exceeds a threshold, however, it can enhance response to the basal inputs that specify the cell's selective sensitivity. This process is referred to as apical amplification (AA). We review evidence suggesting that, by regulating Ih in the apical compartments, adrenergic arousal controls the coupling between apical and somatic integration zones thus modifying cognitive capabilities closely associated with consciousness. Evidence relating AA to schizophrenia, sleep, and anesthesia is reviewed, and we assess theories that emphasize the relevance of AA to consciousness. Implications for theories of neocortical computation that emphasize context-sensitive modulation are summarized. We conclude that the findings concerning AA and its regulation by arousal offer a new perspective on states of consciousness, the function and evolution of neocortex, and psychopathology. Many issues worthy of closer examination arise.
Collapse
Affiliation(s)
- W. A. Phillips
- School of Natural Sciences, University of Stirling, Scotland FK9 4LA, UK
| | - M. E. Larkum
- Neurocure Cluster of Excellence, Department of Biology, Humboldt University,
Charitéplatz 1, Berlin 10117, Germany
| | - C. W. Harley
- Psychology Department, Memorial University of Newfoundland, St. John's, NL A1C 5S7,
P.O. Box 4200, Canada
| | | |
Collapse
|
128
|
Kang E, Wen Z, Song H, Christian KM, Ming GL. Adult Neurogenesis and Psychiatric Disorders. Cold Spring Harb Perspect Biol 2016; 8:cshperspect.a019026. [PMID: 26801682 DOI: 10.1101/cshperspect.a019026] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Psychiatric disorders continue to be among the most challenging disorders to diagnose and treat because there is no single genetic or anatomical locus that is causative for the disease. Current treatments are often blunt tools used to ameliorate the most severe symptoms, at the risk of disrupting functional neural systems. There is a critical need to develop new therapeutic strategies that can target circumscribed functional or anatomical domains of pathology. Adult hippocampal neurogenesis may be one such domain. Here, we review the evidence suggesting that adult hippocampal neurogenesis plays a role in emotional regulation and forms of learning and memory that include temporal and spatial memory encoding and context discrimination, and that its dysregulation is associated with psychiatric disorders, such as affective disorders, schizophrenia, and drug addiction. Further, adult neurogenesis has proven to be an effective model to investigate basic processes of neuronal development and converging evidence suggests that aberrant neural development may be an etiological factor, even in late-onset diseases. Constitutive neurogenesis in the hippocampus of the mature brain reflects large-scale plasticity unique to this region and could be a potential hub for modulation of a subset of cognitive and affective behaviors that are affected by multiple psychiatric disorders.
Collapse
Affiliation(s)
- Eunchai Kang
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Zhexing Wen
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Hongjun Song
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Kimberly M Christian
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Guo-Li Ming
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
129
|
Thomson PA, Duff B, Blackwood DHR, Romaniuk L, Watson A, Whalley HC, Li X, Dauvermann MR, Moorhead TWJ, Bois C, Ryan NM, Redpath H, Hall L, Morris SW, van Beek EJR, Roberts N, Porteous DJ, St Clair D, Whitcher B, Dunlop J, Brandon NJ, Hughes ZA, Hall J, McIntosh A, Lawrie SM. Balanced translocation linked to psychiatric disorder, glutamate, and cortical structure/function. NPJ SCHIZOPHRENIA 2016; 2:16024. [PMID: 27602385 PMCID: PMC4994153 DOI: 10.1038/npjschz.2016.24] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 06/30/2016] [Accepted: 07/01/2016] [Indexed: 01/01/2023]
Abstract
Rare genetic variants of large effect can help elucidate the pathophysiology of brain disorders. Here we expand the clinical and genetic analyses of a family with a (1;11)(q42;q14.3) translocation multiply affected by major psychiatric illness and test the effect of the translocation on the structure and function of prefrontal, and temporal brain regions. The translocation showed significant linkage (LOD score 6.1) with a clinical phenotype that included schizophrenia, schizoaffective disorder, bipolar disorder, and recurrent major depressive disorder. Translocation carriers showed reduced cortical thickness in the left temporal lobe, which correlated with general psychopathology and positive psychotic symptom severity. They showed reduced gyrification in prefrontal cortex, which correlated with general psychopathology severity. Translocation carriers also showed significantly increased activation in the caudate nucleus on increasing verbal working memory load, as well as statistically significant reductions in the right dorsolateral prefrontal cortex glutamate concentrations. These findings confirm that the t(1;11) translocation is associated with a significantly increased risk of major psychiatric disorder and suggest a general vulnerability to psychopathology through altered cortical structure and function, and decreased glutamate levels.
Collapse
Affiliation(s)
- Pippa A Thomson
- Medical Genetics Section, Centre for Genomic and Experimental Medicine, University of Edinburgh, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Western General Hospital , Edinburgh, UK
| | - Barbara Duff
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| | - Douglas H R Blackwood
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| | - Liana Romaniuk
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| | - Andrew Watson
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| | - Heather C Whalley
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| | - Xiang Li
- Clinical Research Imaging Centre (CRIC), The Queen's Medical Research Institute, University of Edinburgh , UK
| | - Maria R Dauvermann
- McGovern Institute for Brain Research, Massachusetts Institute of Technology , Cambridge, MA, USA
| | - T William J Moorhead
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| | - Catherine Bois
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| | - Niamh M Ryan
- Medical Genetics Section, Centre for Genomic and Experimental Medicine, University of Edinburgh, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Western General Hospital , Edinburgh, UK
| | - Holly Redpath
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| | - Lynsey Hall
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| | - Stewart W Morris
- Medical Genetics Section, Centre for Genomic and Experimental Medicine, University of Edinburgh, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Western General Hospital , Edinburgh, UK
| | - Edwin J R van Beek
- Clinical Research Imaging Centre (CRIC), The Queen's Medical Research Institute, University of Edinburgh , UK
| | - Neil Roberts
- Clinical Research Imaging Centre (CRIC), The Queen's Medical Research Institute, University of Edinburgh , UK
| | - David J Porteous
- Medical Genetics Section, Centre for Genomic and Experimental Medicine, University of Edinburgh, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Western General Hospital , Edinburgh, UK
| | - David St Clair
- Institute of Medical Sciences, University of Aberdeen , Aberdeen, UK
| | - Brandon Whitcher
- Clinical & Translational Imaging Group, Pfizer Global Research , Cambridge, MA, USA
| | - John Dunlop
- Neuroscience Research Unit, Pfizer Global Research, Cambridge, MA, USA; AstraZeneca Neuroscience, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Cambridge, MA, USA
| | - Nicholas J Brandon
- Neuroscience Research Unit, Pfizer Global Research, Cambridge, MA, USA; AstraZeneca Neuroscience, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Cambridge, MA, USA
| | - Zoë A Hughes
- Neuroscience Research Unit, Pfizer Global Research , Cambridge, MA, USA
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building , Cardiff, UK
| | - Andrew McIntosh
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| | - Stephen M Lawrie
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| |
Collapse
|
130
|
Piñero-Martos E, Ortega-Vila B, Pol-Fuster J, Cisneros-Barroso E, Ruiz-Guerra L, Medina-Dols A, Heine-Suñer D, Lladó J, Olmos G, Vives-Bauzà C. Disrupted in schizophrenia 1 (DISC1) is a constituent of the mammalian mitochondrial contact site and cristae organizing system (MICOS) complex, and is essential for oxidative phosphorylation. Hum Mol Genet 2016; 25:4157-4169. [PMID: 27466199 DOI: 10.1093/hmg/ddw250] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/18/2016] [Accepted: 07/18/2016] [Indexed: 12/29/2022] Open
Abstract
Disrupted in Schizophrenia-1 (DISC1) has been associated with a broad spectrum of mental disorders. DISC1 is a multi-compartmentalized protein found in the cytoplasm, centrosome, nuclei and mostly enriched in mitochondria. In order to shed light on DISC1 mitochondrial function, we have studied its topology within the organelle. We show in here that in mammals DISC1 resides in the 'Mitochondrial contact site and Cristae Organizing system' (MICOS) complex, involved in cristae organization. DISC1 knockdown in SH-SY5Y cells causes MICOS disassembly and fragmentation of the mitochondrial morphology network. Moreover, DISC1 depleted cells have decreased mitochondrial DNA (mtDNA) content and steady state levels of oxidative phosphorylation (OXPHOS) subunits. As a consequence, OXPHOS complexes and supercomplexes are partially disassembled in DISC1 knockdown cells, which suffer severe bioenergetic defects, evidenced by impaired oxygen consumption, adenosine triphosphate synthesis and mitochondrial membrane potential. Transfection of recombinant full-length human DISC1 restores MICOS complex assembly and rescues OXPHOS function, meanwhile overexpression of the DISC1 truncated form Δ597-854, known to be pathogenic, fails to rescue the bioenergetic impairment caused by DISC1 knockdown. These results should contribute to reveal DISC1 physiological function and potential pathogenic role in severe mental illnesses.
Collapse
Affiliation(s)
- Estefanía Piñero-Martos
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, 07010 Palma de Mallorca, Spain.,Institut d'Investigació Sanitària de Palma (IdISPa), 07010 Palma de Mallorca, Spain
| | - Bernardo Ortega-Vila
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, 07010 Palma de Mallorca, Spain.,Institut d'Investigació Sanitària de Palma (IdISPa), 07010 Palma de Mallorca, Spain
| | - Josep Pol-Fuster
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, 07010 Palma de Mallorca, Spain.,Institut d'Investigació Sanitària de Palma (IdISPa), 07010 Palma de Mallorca, Spain
| | - Eugenia Cisneros-Barroso
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, 07010 Palma de Mallorca, Spain.,Institut d'Investigació Sanitària de Palma (IdISPa), 07010 Palma de Mallorca, Spain
| | - Laura Ruiz-Guerra
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, 07010 Palma de Mallorca, Spain.,Institut d'Investigació Sanitària de Palma (IdISPa), 07010 Palma de Mallorca, Spain
| | - Aina Medina-Dols
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, 07010 Palma de Mallorca, Spain.,Institut d'Investigació Sanitària de Palma (IdISPa), 07010 Palma de Mallorca, Spain
| | - Damián Heine-Suñer
- Institut d'Investigació Sanitària de Palma (IdISPa), 07010 Palma de Mallorca, Spain.,Department of Genetics, Hospital Universitari Son Espases, 07010 Palma de Mallorca, Spain
| | - Jerònia Lladó
- Institut d'Investigació Sanitària de Palma (IdISPa), 07010 Palma de Mallorca, Spain.,Grup de Neurobiologia Cel·lular, Departament de Biologia, i Institut Universitari d'Investigacions en Ciències de la Salut, IUNICS, Universitat de les Illes Balears (UIB), 07122 Palma de Mallorca, Spain
| | - Gabriel Olmos
- Institut d'Investigació Sanitària de Palma (IdISPa), 07010 Palma de Mallorca, Spain.,Grup de Neurobiologia Cel·lular, Departament de Biologia, i Institut Universitari d'Investigacions en Ciències de la Salut, IUNICS, Universitat de les Illes Balears (UIB), 07122 Palma de Mallorca, Spain
| | - Cristofol Vives-Bauzà
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, 07010 Palma de Mallorca, Spain .,Institut d'Investigació Sanitària de Palma (IdISPa), 07010 Palma de Mallorca, Spain
| |
Collapse
|
131
|
Mühle C, Kreczi J, Rhein C, Richter-Schmidinger T, Alexopoulos P, Doerfler A, Lenz B, Kornhuber J. Additive sex-specific influence of common non-synonymous DISC1 variants on amygdala, basal ganglia, and white cortical surface area in healthy young adults. Brain Struct Funct 2016; 222:881-894. [PMID: 27369464 DOI: 10.1007/s00429-016-1253-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 06/16/2016] [Indexed: 01/30/2023]
Abstract
The disrupted-in-schizophrenia-1 (DISC1) gene is known for its role in the development of mental disorders. It is also involved in neurodevelopment, cognition, and memory. To investigate the association between DISC1 variants and brain morphology, we analyzed the influence of the three common non-synonymous polymorphisms in DISC1 on specific brain structures in healthy young adults. The volumes of brain regions were determined in 145 subjects by magnetic resonance imaging and automated analysis using FreeSurfer. Genotyping was performed by high resolution melting of amplified products. In an additive genetic model, rs6675281 (Leu607Phe), rs3738401 (Arg264Gln), and rs821616 (Ser704Cys) significantly explained the volume variance of the amygdala (p = 0.007) and the pallidum (p = 0.004). A higher cumulative portion of minor alleles was associated with larger volumes of the amygdala (p = 0.005), the pallidum (p = 0.001), the caudate (p = 0.024), and the putamen (p = 0.007). Sex-stratified analysis revealed a strong genetic effect of rs6675281 on putamen and pallidum in females but not in males and an opposite influence of rs3738401 on the white cortical surface in females compared to males. The strongest single association was found for rs821616 and the amygdala volume in male subjects (p < 0.001). No effect was detected for the nucleus accumbens. We report-to our knowledge-for the first time a significant and sex-specific influence of common DISC1 variants on volumes of the basal ganglia, the amygdala and on the cortical surface area. Our results demonstrate that the additive model of all three polymorphisms outperforms their single analysis.
Collapse
Affiliation(s)
- Christiane Mühle
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany.
| | - Jakob Kreczi
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Cosima Rhein
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Tanja Richter-Schmidinger
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Panagiotis Alexopoulos
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany.,Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar of the Technical University Munich, Munich, Germany
| | - Arnd Doerfler
- Department of Neuroradiology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Bernd Lenz
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany
| |
Collapse
|
132
|
Jaaro-Peled H, Altimus C, LeGates T, Cash-Padgett T, Zoubovsky S, Hikida T, Ishizuka K, Hattar S, Mongrain V, Sawa A. Abnormal wake/sleep pattern in a novel gain-of-function model of DISC1. Neurosci Res 2016; 112:63-69. [PMID: 27354230 DOI: 10.1016/j.neures.2016.06.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 06/16/2016] [Accepted: 06/20/2016] [Indexed: 01/15/2023]
Abstract
Sleep disturbances are common in psychiatric disorders, but the causal relationship between the two and the underlying genetic factors is unclear. The DISC1 gene is strongly linked to mood disorders and schizophrenia in a Scottish pedigree. In an earlier study we found a sleep homeostasis disturbance in a Drosophila model overexpressing wild-type human DISC1. Here we aimed to explore the relationship between sleep and the DISC1 gene in a mammalian model, a novel transgenic mouse model expressing full-length human DISC1. We assessed circadian rhythms by monitoring wheel running activity under normal 24-h light:dark conditions and in constant darkness and found the DISC1 mice to have normal circadian photoentrainment and normal intrinsic circadian period. We also assessed sleep duration and quality in the DISC1 mice and found that they were awake longer than wild-type controls at baseline with a tendency for lower rebound of delta activity during recovery from a short sleep deprivation. Thus we suggest that DISC1 may be involved in sleep regulation.
Collapse
Affiliation(s)
- Hanna Jaaro-Peled
- Department of Psychiatry, Johns Hopkins University School of Medicine, United States.
| | - Cara Altimus
- Department of Biology, Johns Hopkins University, United States
| | - Tara LeGates
- Department of Biology, Johns Hopkins University, United States
| | - Tyler Cash-Padgett
- Department of Psychiatry, Johns Hopkins University School of Medicine, United States
| | - Sandra Zoubovsky
- Department of Psychiatry, Johns Hopkins University School of Medicine, United States
| | - Takatoshi Hikida
- Department of Psychiatry, Johns Hopkins University School of Medicine, United States
| | - Koko Ishizuka
- Department of Psychiatry, Johns Hopkins University School of Medicine, United States
| | - Samer Hattar
- Department of Biology, Johns Hopkins University, United States.
| | | | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, United States.
| |
Collapse
|
133
|
Borkowska M, Millar JK, Price DJ. Altered Disrupted-in-Schizophrenia-1 Function Affects the Development of Cortical Parvalbumin Interneurons by an Indirect Mechanism. PLoS One 2016; 11:e0156082. [PMID: 27244370 PMCID: PMC4886955 DOI: 10.1371/journal.pone.0156082] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 05/09/2016] [Indexed: 01/01/2023] Open
Abstract
Disrupted-in-Schizophrenia-1 (DISC1) gene has been linked to schizophrenia and related major mental illness. Mouse Disc1 has been implicated in brain development, mainly in the proliferation, differentiation, lamination, neurite outgrowth and synapse formation and maintenance of cortical excitatory neurons. Here, the effects of two loss-of-function point mutations in the mouse Disc1 sequence (Q31L and L100P) on cortical inhibitory interneurons were investigated. None of the mutations affected the overall number of interneurons. However, the 100P, but not the 31L, mutation resulted in a significant decrease in the numbers of interneurons expressing parvalbumin mRNA and protein across the sensory cortex. To investigate role of Disc1 in regulation of parvalbumin expression, mouse wild-type Disc-1 or the 100P mutant form were electroporated in utero into cortical excitatory neurons. Overexpression of wild-type Disc1 in these cells caused increased densities of parvalbumin-expressing interneurons in the electroporated area and in areas connected with it, whereas expression of Disc1-100P did not. We conclude that the 100P mutation prevents expression of parvalbumin by a normally sized cohort of interneurons and that altering Disc1 function in cortical excitatory neurons indirectly affects parvalbumin expression by cortical interneurons, perhaps as a result of altered functional input from the excitatory neurons.
Collapse
Affiliation(s)
- Malgorzata Borkowska
- University of Edinburgh Centre for Integrative Physiology, Hugh Robson Building, George Square, Edinburgh EH8 9XD, United Kingdom
- * E-mail:
| | - J. Kirsty Millar
- University of Edinburgh Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Crewe Road, Edinburgh EH4 2XU, United Kingdom
| | - David J. Price
- University of Edinburgh Centre for Integrative Physiology, Hugh Robson Building, George Square, Edinburgh EH8 9XD, United Kingdom
| |
Collapse
|
134
|
Ogawa F, Murphy LC, Malavasi ELV, O’Sullivan ST, Torrance HS, Porteous DJ, Millar JK. NDE1 and GSK3β Associate with TRAK1 and Regulate Axonal Mitochondrial Motility: Identification of Cyclic AMP as a Novel Modulator of Axonal Mitochondrial Trafficking. ACS Chem Neurosci 2016; 7:553-64. [PMID: 26815013 DOI: 10.1021/acschemneuro.5b00255] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Mitochondria are essential for neuronal function, providing the energy required to power neurotransmission, and fulfilling many important additional roles. In neurons, mitochondria must be efficiently transported to sites, including synapses, where their functions are required. Neurons, with their highly elongated morphology, are consequently extremely sensitive to defective mitochondrial trafficking which can lead to neuronal ill-health/death. We recently demonstrated that DISC1 associates with mitochondrial trafficking complexes where it associates with the core kinesin and dynein adaptor molecule TRAK1. We now show that the DISC1 interactors NDE1 and GSK3β also associate robustly with TRAK1 and demonstrate that NDE1 promotes retrograde axonal mitochondrial movement. GSK3β is known to modulate axonal mitochondrial motility, although reports of its actual effect are conflicting. We show that, in our system, GSK3β promotes anterograde mitochondrial transport. Finally, we investigated the influence of cAMP elevation upon mitochondrial motility, and found a striking increase in mitochondrial motility and retrograde movement. DISC1, NDE1, and GSK3β are implicated as risk factors for major mental illness. Our demonstration that they function together within mitochondrial trafficking complexes suggests that defective mitochondrial transport may be a contributory disease mechanism in some cases of psychiatric disorder.
Collapse
Affiliation(s)
- Fumiaki Ogawa
- University
of Edinburgh Centre
for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Crewe Road, Edinburgh, EH4 2XU, United Kingdom
| | - Laura C. Murphy
- University
of Edinburgh Centre
for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Crewe Road, Edinburgh, EH4 2XU, United Kingdom
| | - Elise L. V. Malavasi
- University
of Edinburgh Centre
for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Crewe Road, Edinburgh, EH4 2XU, United Kingdom
| | - Shane T. O’Sullivan
- University
of Edinburgh Centre
for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Crewe Road, Edinburgh, EH4 2XU, United Kingdom
| | - Helen S. Torrance
- University
of Edinburgh Centre
for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Crewe Road, Edinburgh, EH4 2XU, United Kingdom
| | - David J. Porteous
- University
of Edinburgh Centre
for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Crewe Road, Edinburgh, EH4 2XU, United Kingdom
| | - J. Kirsty Millar
- University
of Edinburgh Centre
for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Crewe Road, Edinburgh, EH4 2XU, United Kingdom
| |
Collapse
|
135
|
Tomoda T, Sumitomo A, Jaaro-Peled H, Sawa A. Utility and validity of DISC1 mouse models in biological psychiatry. Neuroscience 2016; 321:99-107. [PMID: 26768401 PMCID: PMC4803604 DOI: 10.1016/j.neuroscience.2015.12.061] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 12/31/2015] [Accepted: 12/31/2015] [Indexed: 11/26/2022]
Abstract
We have seen an era of explosive progress in translating neurobiology into etiological understanding of mental disorders for the past 10-15 years. The discovery of Disrupted-in-schizophrenia 1 (DISC1) gene was one of the major driving forces that have contributed to the progress. The finding that DISC1 plays crucial roles in neurodevelopment and synapse regulation clearly underscored the utility and validity of DISC1-related biology in advancing our understanding of pathophysiological processes underlying psychiatric conditions. Despite recent genetic studies that failed to identify DISC1 as a risk gene for sporadic cases of schizophrenia, DISC1 mutant mice, coupled with various environmental stressors, have proven successful in satisfying face validity as models of a wide range of human psychiatric conditions. Investigating mental disorders using these models is expected to further contribute to the circuit-level understanding of the pathological mechanisms, as well as to the development of novel therapeutic strategies in the future.
Collapse
Affiliation(s)
- T Tomoda
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| | - A Sumitomo
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - H Jaaro-Peled
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - A Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
136
|
Chen J, He K, Wang Q, Li Z, Shen J, Li T, Wang M, Wen Z, Li W, Qiang Y, Wang T, Ji J, Wu N, Wang Z, Zhang B, Feng G, He L, Xu Y, Shi Y. Role played by the SP4 gene in schizophrenia and major depressive disorder in the Han Chinese population. Br J Psychiatry 2016; 208:441-445. [PMID: 26450579 DOI: 10.1192/bjp.bp.114.151688] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Accepted: 11/07/2014] [Indexed: 01/27/2023]
Abstract
BACKGROUND Psychiatric disorders such as schizophrenia and major depressive disorder (MDD) are likely to be caused by multiple susceptibility genes, each with small effects in increasing the risk of illness. Identifying DNA variants associated with schizophrenia and MDD is a crucial step in understanding the pathophysiology of these disorders. AIMS To investigate whether the SP4 gene plays a significant role in schizophrenia or MDD in the Han Chinese population. METHOD We focused on nine single nucleotide polymorphisms (SNPs) harbouring the SP4 gene and carried out case-control studies in 1235 patients with schizophrenia, 1045 patients with MDD and 1235 healthy controls recruited from the Han Chinese population. RESULTS We found that rs40245 was significantly associated with schizophrenia in both allele and genotype distributions (Pallele = 0.0005, Pallele = 0.004 after Bonferroni correction; Pgenotype = 0.0023, Pgenotype = 0.0184 after Bonferroni correction). The rs6461563 SNP was significantly associated with schizophrenia in the allele distributions (Pallele = 0.0033, Pallele = 0.0264 after Bonferroni correction). CONCLUSIONS Our results suggest that common risk factors in the SP4 gene are associated with schizophrenia, although not with MDD, in the Han Chinese population.
Collapse
Affiliation(s)
- Jianhua Chen
- Jianhua Chen, MD, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Kuanjun He, PhD, Qingzhong Wang, PhD, Zhiqiang Li, PhD, Jiawei Shen, PhD, Tao Li, PhD, Meng Wang, MSc, Zujia Wen, PhD, Wenjin Li, PhD, Yu Qiang, MSc, Ti Wang, PhD, Jue Ji, BS, Na Wu, BS, Zhiqiao Wang, BS, Bo Zhang, BS, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Guoyin Feng, BS, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Lin He, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Yifeng Xu, MD, MSc, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Yongyong Shi, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, Shanghai Changning Mental Health Center, Shanghai, Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, and Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Kuanjun He
- Jianhua Chen, MD, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Kuanjun He, PhD, Qingzhong Wang, PhD, Zhiqiang Li, PhD, Jiawei Shen, PhD, Tao Li, PhD, Meng Wang, MSc, Zujia Wen, PhD, Wenjin Li, PhD, Yu Qiang, MSc, Ti Wang, PhD, Jue Ji, BS, Na Wu, BS, Zhiqiao Wang, BS, Bo Zhang, BS, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Guoyin Feng, BS, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Lin He, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Yifeng Xu, MD, MSc, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Yongyong Shi, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, Shanghai Changning Mental Health Center, Shanghai, Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, and Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Qingzhong Wang
- Jianhua Chen, MD, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Kuanjun He, PhD, Qingzhong Wang, PhD, Zhiqiang Li, PhD, Jiawei Shen, PhD, Tao Li, PhD, Meng Wang, MSc, Zujia Wen, PhD, Wenjin Li, PhD, Yu Qiang, MSc, Ti Wang, PhD, Jue Ji, BS, Na Wu, BS, Zhiqiao Wang, BS, Bo Zhang, BS, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Guoyin Feng, BS, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Lin He, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Yifeng Xu, MD, MSc, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Yongyong Shi, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, Shanghai Changning Mental Health Center, Shanghai, Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, and Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiqiang Li
- Jianhua Chen, MD, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Kuanjun He, PhD, Qingzhong Wang, PhD, Zhiqiang Li, PhD, Jiawei Shen, PhD, Tao Li, PhD, Meng Wang, MSc, Zujia Wen, PhD, Wenjin Li, PhD, Yu Qiang, MSc, Ti Wang, PhD, Jue Ji, BS, Na Wu, BS, Zhiqiao Wang, BS, Bo Zhang, BS, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Guoyin Feng, BS, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Lin He, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Yifeng Xu, MD, MSc, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Yongyong Shi, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, Shanghai Changning Mental Health Center, Shanghai, Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, and Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Jiawei Shen
- Jianhua Chen, MD, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Kuanjun He, PhD, Qingzhong Wang, PhD, Zhiqiang Li, PhD, Jiawei Shen, PhD, Tao Li, PhD, Meng Wang, MSc, Zujia Wen, PhD, Wenjin Li, PhD, Yu Qiang, MSc, Ti Wang, PhD, Jue Ji, BS, Na Wu, BS, Zhiqiao Wang, BS, Bo Zhang, BS, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Guoyin Feng, BS, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Lin He, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Yifeng Xu, MD, MSc, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Yongyong Shi, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, Shanghai Changning Mental Health Center, Shanghai, Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, and Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Li
- Jianhua Chen, MD, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Kuanjun He, PhD, Qingzhong Wang, PhD, Zhiqiang Li, PhD, Jiawei Shen, PhD, Tao Li, PhD, Meng Wang, MSc, Zujia Wen, PhD, Wenjin Li, PhD, Yu Qiang, MSc, Ti Wang, PhD, Jue Ji, BS, Na Wu, BS, Zhiqiao Wang, BS, Bo Zhang, BS, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Guoyin Feng, BS, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Lin He, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Yifeng Xu, MD, MSc, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Yongyong Shi, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, Shanghai Changning Mental Health Center, Shanghai, Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, and Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Meng Wang
- Jianhua Chen, MD, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Kuanjun He, PhD, Qingzhong Wang, PhD, Zhiqiang Li, PhD, Jiawei Shen, PhD, Tao Li, PhD, Meng Wang, MSc, Zujia Wen, PhD, Wenjin Li, PhD, Yu Qiang, MSc, Ti Wang, PhD, Jue Ji, BS, Na Wu, BS, Zhiqiao Wang, BS, Bo Zhang, BS, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Guoyin Feng, BS, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Lin He, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Yifeng Xu, MD, MSc, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Yongyong Shi, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, Shanghai Changning Mental Health Center, Shanghai, Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, and Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Zujia Wen
- Jianhua Chen, MD, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Kuanjun He, PhD, Qingzhong Wang, PhD, Zhiqiang Li, PhD, Jiawei Shen, PhD, Tao Li, PhD, Meng Wang, MSc, Zujia Wen, PhD, Wenjin Li, PhD, Yu Qiang, MSc, Ti Wang, PhD, Jue Ji, BS, Na Wu, BS, Zhiqiao Wang, BS, Bo Zhang, BS, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Guoyin Feng, BS, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Lin He, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Yifeng Xu, MD, MSc, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Yongyong Shi, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, Shanghai Changning Mental Health Center, Shanghai, Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, and Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Wenjin Li
- Jianhua Chen, MD, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Kuanjun He, PhD, Qingzhong Wang, PhD, Zhiqiang Li, PhD, Jiawei Shen, PhD, Tao Li, PhD, Meng Wang, MSc, Zujia Wen, PhD, Wenjin Li, PhD, Yu Qiang, MSc, Ti Wang, PhD, Jue Ji, BS, Na Wu, BS, Zhiqiao Wang, BS, Bo Zhang, BS, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Guoyin Feng, BS, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Lin He, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Yifeng Xu, MD, MSc, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Yongyong Shi, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, Shanghai Changning Mental Health Center, Shanghai, Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, and Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Qiang
- Jianhua Chen, MD, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Kuanjun He, PhD, Qingzhong Wang, PhD, Zhiqiang Li, PhD, Jiawei Shen, PhD, Tao Li, PhD, Meng Wang, MSc, Zujia Wen, PhD, Wenjin Li, PhD, Yu Qiang, MSc, Ti Wang, PhD, Jue Ji, BS, Na Wu, BS, Zhiqiao Wang, BS, Bo Zhang, BS, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Guoyin Feng, BS, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Lin He, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Yifeng Xu, MD, MSc, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Yongyong Shi, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, Shanghai Changning Mental Health Center, Shanghai, Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, and Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Ti Wang
- Jianhua Chen, MD, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Kuanjun He, PhD, Qingzhong Wang, PhD, Zhiqiang Li, PhD, Jiawei Shen, PhD, Tao Li, PhD, Meng Wang, MSc, Zujia Wen, PhD, Wenjin Li, PhD, Yu Qiang, MSc, Ti Wang, PhD, Jue Ji, BS, Na Wu, BS, Zhiqiao Wang, BS, Bo Zhang, BS, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Guoyin Feng, BS, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Lin He, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Yifeng Xu, MD, MSc, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Yongyong Shi, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, Shanghai Changning Mental Health Center, Shanghai, Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, and Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Jue Ji
- Jianhua Chen, MD, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Kuanjun He, PhD, Qingzhong Wang, PhD, Zhiqiang Li, PhD, Jiawei Shen, PhD, Tao Li, PhD, Meng Wang, MSc, Zujia Wen, PhD, Wenjin Li, PhD, Yu Qiang, MSc, Ti Wang, PhD, Jue Ji, BS, Na Wu, BS, Zhiqiao Wang, BS, Bo Zhang, BS, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Guoyin Feng, BS, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Lin He, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Yifeng Xu, MD, MSc, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Yongyong Shi, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, Shanghai Changning Mental Health Center, Shanghai, Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, and Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Na Wu
- Jianhua Chen, MD, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Kuanjun He, PhD, Qingzhong Wang, PhD, Zhiqiang Li, PhD, Jiawei Shen, PhD, Tao Li, PhD, Meng Wang, MSc, Zujia Wen, PhD, Wenjin Li, PhD, Yu Qiang, MSc, Ti Wang, PhD, Jue Ji, BS, Na Wu, BS, Zhiqiao Wang, BS, Bo Zhang, BS, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Guoyin Feng, BS, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Lin He, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Yifeng Xu, MD, MSc, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Yongyong Shi, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, Shanghai Changning Mental Health Center, Shanghai, Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, and Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiqiao Wang
- Jianhua Chen, MD, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Kuanjun He, PhD, Qingzhong Wang, PhD, Zhiqiang Li, PhD, Jiawei Shen, PhD, Tao Li, PhD, Meng Wang, MSc, Zujia Wen, PhD, Wenjin Li, PhD, Yu Qiang, MSc, Ti Wang, PhD, Jue Ji, BS, Na Wu, BS, Zhiqiao Wang, BS, Bo Zhang, BS, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Guoyin Feng, BS, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Lin He, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Yifeng Xu, MD, MSc, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Yongyong Shi, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, Shanghai Changning Mental Health Center, Shanghai, Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, and Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Zhang
- Jianhua Chen, MD, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Kuanjun He, PhD, Qingzhong Wang, PhD, Zhiqiang Li, PhD, Jiawei Shen, PhD, Tao Li, PhD, Meng Wang, MSc, Zujia Wen, PhD, Wenjin Li, PhD, Yu Qiang, MSc, Ti Wang, PhD, Jue Ji, BS, Na Wu, BS, Zhiqiao Wang, BS, Bo Zhang, BS, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Guoyin Feng, BS, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Lin He, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Yifeng Xu, MD, MSc, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Yongyong Shi, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, Shanghai Changning Mental Health Center, Shanghai, Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, and Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Guoyin Feng
- Jianhua Chen, MD, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Kuanjun He, PhD, Qingzhong Wang, PhD, Zhiqiang Li, PhD, Jiawei Shen, PhD, Tao Li, PhD, Meng Wang, MSc, Zujia Wen, PhD, Wenjin Li, PhD, Yu Qiang, MSc, Ti Wang, PhD, Jue Ji, BS, Na Wu, BS, Zhiqiao Wang, BS, Bo Zhang, BS, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Guoyin Feng, BS, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Lin He, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Yifeng Xu, MD, MSc, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Yongyong Shi, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, Shanghai Changning Mental Health Center, Shanghai, Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, and Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Lin He
- Jianhua Chen, MD, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Kuanjun He, PhD, Qingzhong Wang, PhD, Zhiqiang Li, PhD, Jiawei Shen, PhD, Tao Li, PhD, Meng Wang, MSc, Zujia Wen, PhD, Wenjin Li, PhD, Yu Qiang, MSc, Ti Wang, PhD, Jue Ji, BS, Na Wu, BS, Zhiqiao Wang, BS, Bo Zhang, BS, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Guoyin Feng, BS, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Lin He, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Yifeng Xu, MD, MSc, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Yongyong Shi, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, Shanghai Changning Mental Health Center, Shanghai, Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, and Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Yifeng Xu
- Jianhua Chen, MD, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Kuanjun He, PhD, Qingzhong Wang, PhD, Zhiqiang Li, PhD, Jiawei Shen, PhD, Tao Li, PhD, Meng Wang, MSc, Zujia Wen, PhD, Wenjin Li, PhD, Yu Qiang, MSc, Ti Wang, PhD, Jue Ji, BS, Na Wu, BS, Zhiqiao Wang, BS, Bo Zhang, BS, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Guoyin Feng, BS, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Lin He, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Yifeng Xu, MD, MSc, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Yongyong Shi, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, Shanghai Changning Mental Health Center, Shanghai, Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, and Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Yongyong Shi
- Jianhua Chen, MD, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Kuanjun He, PhD, Qingzhong Wang, PhD, Zhiqiang Li, PhD, Jiawei Shen, PhD, Tao Li, PhD, Meng Wang, MSc, Zujia Wen, PhD, Wenjin Li, PhD, Yu Qiang, MSc, Ti Wang, PhD, Jue Ji, BS, Na Wu, BS, Zhiqiao Wang, BS, Bo Zhang, BS, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Guoyin Feng, BS, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Lin He, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China; Yifeng Xu, MD, MSc, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Yongyong Shi, PhD, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, Shanghai Changning Mental Health Center, Shanghai, Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, and Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
137
|
Impairments in dendrite morphogenesis as etiology for neurodevelopmental disorders and implications for therapeutic treatments. Neurosci Biobehav Rev 2016; 68:946-978. [PMID: 27143622 DOI: 10.1016/j.neubiorev.2016.04.008] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 04/13/2016] [Accepted: 04/13/2016] [Indexed: 02/08/2023]
Abstract
Dendrite morphology is pivotal for neural circuitry functioning. While the causative relationship between small-scale dendrite morphological abnormalities (shape, density of dendritic spines) and neurodevelopmental disorders is well established, such relationship remains elusive for larger-scale dendrite morphological impairments (size, shape, branching pattern of dendritic trees). Here, we summarize published data on dendrite morphological irregularities in human patients and animal models for neurodevelopmental disorders, with focus on autism and schizophrenia. We next discuss high-risk genes for these disorders and their role in dendrite morphogenesis. We finally overview recent developments in therapeutic attempts and we discuss how they relate to dendrite morphology. We find that both autism and schizophrenia are accompanied by dendritic arbor morphological irregularities, and that majority of their high-risk genes regulate dendrite morphogenesis. Thus, we present a compelling argument that, along with smaller-scale morphological impairments in dendrites (spines and synapse), irregularities in larger-scale dendrite morphology (arbor shape, size) may be an important part of neurodevelopmental disorders' etiology. We suggest that this should not be ignored when developing future therapeutic treatments.
Collapse
|
138
|
O'Tuathaigh CMP, Desbonnet L, Moran PM, Kirby BP, Waddington JL. Molecular genetic models related to schizophrenia and psychotic illness: heuristics and challenges. Curr Top Behav Neurosci 2016; 7:87-119. [PMID: 21298380 DOI: 10.1007/7854_2010_111] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Schizophrenia is a heritable disorder that may involve several common genes of small effect and/or rare copy number variation, with phenotypic heterogeneity across patients. Furthermore, any boundaries vis-à-vis other psychotic disorders are far from clear. Consequently, identification of informative animal models for this disorder, which typically relate to pharmacological and putative pathophysiological processes of uncertain validity, faces considerable challenges. In juxtaposition, the majority of mutant models for schizophrenia relate to the functional roles of a diverse set of genes associated with risk for the disorder or with such putative pathophysiological processes. This chapter seeks to outline the evidence from phenotypic studies in mutant models related to schizophrenia. These have commonly assessed the degree to which mutation of a schizophrenia-related gene is associated with the expression of several aspects of the schizophrenia phenotype or more circumscribed, schizophrenia-related endophenotypes; typically, they place specific emphasis on positive and negative symptoms and cognitive deficits, and extend to structural and other pathological features. We first consider the primary technological approaches to the generation of such mutants, to include their relative merits and demerits, and then highlight the diverse phenotypic approaches that have been developed for their assessment. The chapter then considers the application of mutant phenotypes to study pathobiological and pharmacological mechanisms thought to be relevant for schizophrenia, particularly in terms of dopaminergic and glutamatergic dysfunction, and to an increasing range of candidate susceptibility genes and copy number variants. Finally, we discuss several pertinent issues and challenges within the field which relate to both phenotypic evaluation and a growing appreciation of the functional genomics of schizophrenia and the involvement of gene × environment interactions.
Collapse
Affiliation(s)
- Colm M P O'Tuathaigh
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland,
| | | | | | | | | |
Collapse
|
139
|
McGirr A, Lipina TV, Mun HS, Georgiou J, Al-Amri AH, Ng E, Zhai D, Elliott C, Cameron RT, Mullins JGL, Liu F, Baillie GS, Clapcote SJ, Roder JC. Specific Inhibition of Phosphodiesterase-4B Results in Anxiolysis and Facilitates Memory Acquisition. Neuropsychopharmacology 2016; 41:1080-92. [PMID: 26272049 PMCID: PMC4748432 DOI: 10.1038/npp.2015.240] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 07/30/2015] [Accepted: 08/03/2015] [Indexed: 01/07/2023]
Abstract
Cognitive dysfunction is a core feature of dementia and a prominent feature in psychiatric disease. As non-redundant regulators of intracellular cAMP gradients, phosphodiesterases (PDE) mediate fundamental aspects of brain function relevant to learning, memory, and higher cognitive functions. Phosphodiesterase-4B (PDE4B) is an important phosphodiesterase in the hippocampal formation, is a major Disrupted in Schizophrenia 1 (DISC1) binding partner and is itself a risk gene for psychiatric illness. To define the effects of specific inhibition of the PDE4B subtype, we generated mice with a catalytic domain mutant form of PDE4B (Y358C) that has decreased ability to hydrolyze cAMP. Structural modeling predictions of decreased function and impaired binding with DISC1 were confirmed in cell assays. Phenotypic characterization of the PDE4B(Y358C) mice revealed facilitated phosphorylation of CREB, decreased binding to DISC1, and upregulation of DISC1 and β-Arrestin in hippocampus and amygdala. In behavioral assays, PDE4B(Y358C) mice displayed decreased anxiety and increased exploration, as well as cognitive enhancement across several tests of learning and memory, consistent with synaptic changes including enhanced long-term potentiation and impaired depotentiation ex vivo. PDE4B(Y358C) mice also demonstrated enhanced neurogenesis. Contextual fear memory, though intact at 24 h, was decreased at 7 days in PDE4B(Y358C) mice, an effect replicated pharmacologically with a non-selective PDE4 inhibitor, implicating cAMP signaling by PDE4B in a very late phase of consolidation. No effect of the PDE4B(Y358C) mutation was observed in the prepulse inhibition and forced swim tests. Our data establish specific inhibition of PDE4B as a promising therapeutic approach for disorders of cognition and anxiety, and a putative target for pathological fear memory.
Collapse
Affiliation(s)
- Alexander McGirr
- Department of Psychiatry, University of
British Columbia, Vancouver, British Columbia,
Canada,Lunenfeld-Tanenbaum Research Institute,
Mount Sinai Hospital, Toronto, Ontario,
Canada,Department of Psychiatry, University of British
Columbia, Vancouver, British Columbia,
Canada
V6T 2A1, E-mail:
| | - Tatiana V Lipina
- Lunenfeld-Tanenbaum Research Institute,
Mount Sinai Hospital, Toronto, Ontario,
Canada
| | - Ho-Suk Mun
- Lunenfeld-Tanenbaum Research Institute,
Mount Sinai Hospital, Toronto, Ontario,
Canada,Department of Medical Genetics,
University of Toronto, Toronto, Ontario,
Canada
| | - John Georgiou
- Lunenfeld-Tanenbaum Research Institute,
Mount Sinai Hospital, Toronto, Ontario,
Canada
| | - Ahmed H Al-Amri
- School of Biomedical Sciences, University
of Leeds, Leeds, UK,National Genetic Centre, Royal
Hospital, Muscat, Oman
| | - Enoch Ng
- Lunenfeld-Tanenbaum Research Institute,
Mount Sinai Hospital, Toronto, Ontario,
Canada,Institute of Medical Science, University
of Toronto, Toronto, Ontario,
Canada
| | - Dongxu Zhai
- Department of Neuroscience, Centre for
Addiction and Mental Health, Toronto, Ontario,
Canada
| | - Christina Elliott
- Institute of Cardiovascular and Medical
Sciences, College of Medical, Veterinary and Life Sciences, University of
Glasgow, Glasgow, UK
| | - Ryan T Cameron
- Institute of Cardiovascular and Medical
Sciences, College of Medical, Veterinary and Life Sciences, University of
Glasgow, Glasgow, UK
| | - Jonathan GL Mullins
- Institute of Life Science, College of
Medicine, Swansea University, Swansea, UK
| | - Fang Liu
- Department of Neuroscience, Centre for
Addiction and Mental Health, Toronto, Ontario,
Canada
| | - George S Baillie
- Institute of Cardiovascular and Medical
Sciences, College of Medical, Veterinary and Life Sciences, University of
Glasgow, Glasgow, UK
| | - Steven J Clapcote
- School of Biomedical Sciences, University
of Leeds, Leeds, UK,School of Biomedical Sciences, University of Leeds,
Leeds
LS2 9JT, UK, Tel: +44 (0)113 3433041,
E-mail:
| | - John C Roder
- Lunenfeld-Tanenbaum Research Institute,
Mount Sinai Hospital, Toronto, Ontario,
Canada,Department of Physiology, University of
Toronto, Toronto, Ontario, Canada
| |
Collapse
|
140
|
Brownstein CA, Kleiman RJ, Engle EC, Towne MC, D'Angelo EJ, Yu TW, Beggs AH, Picker J, Fogler JM, Carroll D, Schmitt RCO, Wolff RR, Shen Y, Lip V, Bilguvar K, Kim A, Tembulkar S, O'Donnell K, Gonzalez-Heydrich J. Overlapping 16p13.11 deletion and gain of copies variations associated with childhood onset psychosis include genes with mechanistic implications for autism associated pathways: Two case reports. Am J Med Genet A 2016; 170A:1165-73. [PMID: 26887912 PMCID: PMC4833544 DOI: 10.1002/ajmg.a.37595] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 01/16/2016] [Indexed: 12/15/2022]
Abstract
Copy number variability at 16p13.11 has been associated with intellectual disability, autism, schizophrenia, epilepsy, and attention-deficit hyperactivity disorder. Adolescent/adult- onset psychosis has been reported in a subset of these cases. Here, we report on two children with CNVs in 16p13.11 that developed psychosis before the age of 7. The genotype and neuropsychiatric abnormalities of these patients highlight several overlapping genes that have possible mechanistic relevance to pathways previously implicated in Autism Spectrum Disorders, including the mTOR signaling and the ubiquitin-proteasome cascades. A careful screening of the 16p13.11 region is warranted in patients with childhood onset psychosis.
Collapse
Affiliation(s)
- Catherine A Brownstein
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts.,Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Robin J Kleiman
- Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts.,FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts.,Department of Neurology, Harvard Medical School, Boston, Massachusetts.,Department of Neurology, Boston Children's Hospital, Boston, Massachusetts
| | - Elizabeth C Engle
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts.,Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts.,FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts.,Department of Neurology, Harvard Medical School, Boston, Massachusetts.,Department of Neurology, Boston Children's Hospital, Boston, Massachusetts.,Department of Ophthalmology, Boston Children's Hospital, Boston, Massachusetts.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts.,Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Meghan C Towne
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts.,Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts
| | - Eugene J D'Angelo
- Division of Psychology, Department of Psychiatry, Boston Children's Hospital, Boston, Massachusetts.,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts.,Department of Psychiatry, Developmental Neuropsychiatry Research Program, Boston Children's Hospital, Boston, Massachusetts
| | - Timothy W Yu
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts.,Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Alan H Beggs
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts.,Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Jonathan Picker
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts.,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Jason M Fogler
- Division of Psychology, Department of Psychiatry, Boston Children's Hospital, Boston, Massachusetts.,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts.,Developmental Medicine Center, Boston Children's Hospital, Boston, Massachusetts
| | - Devon Carroll
- Department of Psychiatry, Developmental Neuropsychiatry Research Program, Boston Children's Hospital, Boston, Massachusetts
| | - Rachel C O Schmitt
- Department of Neurology, Boston Children's Hospital, Boston, Massachusetts.,Division of Psychology, Department of Psychiatry, Boston Children's Hospital, Boston, Massachusetts.,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Robert R Wolff
- Department of Neurology, Harvard Medical School, Boston, Massachusetts.,Department of Neurology, Boston Children's Hospital, Boston, Massachusetts
| | - Yiping Shen
- Claritas Genomics, Cambridge, Massachusetts.,Department of Laboratory Medicine, Boston Children's Hospital, Boston, Massachusetts.,Department of Genetics, Yale Center for Genome Analysis, Yale School of Medicine, New Haven, Connecticut
| | - Va Lip
- Claritas Genomics, Cambridge, Massachusetts
| | - Kaya Bilguvar
- Department of Pathology, Harvard Medical School, Boston, Massachusetts
| | - April Kim
- Department of Psychiatry, Developmental Neuropsychiatry Research Program, Boston Children's Hospital, Boston, Massachusetts
| | - Sahil Tembulkar
- Department of Psychiatry, Developmental Neuropsychiatry Research Program, Boston Children's Hospital, Boston, Massachusetts
| | - Kyle O'Donnell
- Department of Psychiatry, Developmental Neuropsychiatry Research Program, Boston Children's Hospital, Boston, Massachusetts
| | - Joseph Gonzalez-Heydrich
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts.,Department of Psychiatry, Developmental Neuropsychiatry Research Program, Boston Children's Hospital, Boston, Massachusetts
| |
Collapse
|
141
|
Sawa A, Ishizuka K, Katsanis N. The potential of DISC1 protein as a therapeutic target for mental illness. Expert Opin Ther Targets 2016; 20:641-3. [PMID: 26810812 DOI: 10.1517/14728222.2016.1146694] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Akira Sawa
- a Department of Psychiatry and Behavioral Sciences , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Koko Ishizuka
- a Department of Psychiatry and Behavioral Sciences , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Nicholas Katsanis
- b Center for Human Disease Modeling and Departments of Cell Biology and Pediatrics , Duke University School of Medicine , Durham , NC , USA
| |
Collapse
|
142
|
Wang Q, Amato SP, Rubitski DM, Hayward MM, Kormos BL, Verhoest PR, Xu L, Brandon NJ, Ehlers MD. Identification of Phosphorylation Consensus Sequences and Endogenous Neuronal Substrates of the Psychiatric Risk Kinase TNIK. J Pharmacol Exp Ther 2016; 356:410-23. [PMID: 26645429 DOI: 10.1124/jpet.115.229880] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/01/2015] [Indexed: 12/28/2022] Open
Abstract
Traf2- and Nck-interacting kinase (TNIK) is a serine/threonine kinase highly expressed in the brain and enriched in the postsynaptic density of glutamatergic synapses in the mammalian brain. Accumulating genetic evidence and functional data have implicated TNIK as a risk factor for psychiatric disorders. However, the endogenous substrates of TNIK in neurons are unknown. Here, we describe a novel selective small molecule inhibitor of the TNIK kinase family. Using this inhibitor, we report the identification of endogenous neuronal TNIK substrates by immunoprecipitation with a phosphomotif antibody followed by mass spectrometry. Phosphorylation consensus sequences were defined by phosphopeptide sequence analysis. Among the identified substrates were members of the delta-catenin family including p120-catenin, δ-catenin, and armadillo repeat gene deleted in velo-cardio-facial syndrome (ARVCF), each of which is linked to psychiatric or neurologic disorders. Using p120-catenin as a representative substrate, we show TNIK-induced p120-catenin phosphorylation in cells requires intact kinase activity and phosphorylation of TNIK at T181 and T187 in the activation loop. Addition of the small molecule TNIK inhibitor or knocking down TNIK by two shRNAs reduced endogenous p120-catenin phosphorylation in cells. Together, using a TNIK inhibitor and phosphomotif antibody, we identify endogenous substrates of TNIK in neurons, define consensus sequences for TNIK, and suggest signaling pathways by which TNIK influences synaptic development and function linked to psychiatric and neurologic disorders.
Collapse
Affiliation(s)
- Qi Wang
- Neuroscience & Pain Research Unit, BioTherapeutics Research and Development, Pfizer Inc. Cambridge, Massachusetts (Q.W., S.P.A., D.M.R., N.J.B., M.D.E.); Center of Chemistry Innovation and Excellence, Pfizer Inc., Groton, Connecticut (M.M.H.); Neuroscience Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts (B.L.K., P.R.V.);and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts (L.X.)
| | - Stephen P Amato
- Neuroscience & Pain Research Unit, BioTherapeutics Research and Development, Pfizer Inc. Cambridge, Massachusetts (Q.W., S.P.A., D.M.R., N.J.B., M.D.E.); Center of Chemistry Innovation and Excellence, Pfizer Inc., Groton, Connecticut (M.M.H.); Neuroscience Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts (B.L.K., P.R.V.);and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts (L.X.)
| | - David M Rubitski
- Neuroscience & Pain Research Unit, BioTherapeutics Research and Development, Pfizer Inc. Cambridge, Massachusetts (Q.W., S.P.A., D.M.R., N.J.B., M.D.E.); Center of Chemistry Innovation and Excellence, Pfizer Inc., Groton, Connecticut (M.M.H.); Neuroscience Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts (B.L.K., P.R.V.);and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts (L.X.)
| | - Matthew M Hayward
- Neuroscience & Pain Research Unit, BioTherapeutics Research and Development, Pfizer Inc. Cambridge, Massachusetts (Q.W., S.P.A., D.M.R., N.J.B., M.D.E.); Center of Chemistry Innovation and Excellence, Pfizer Inc., Groton, Connecticut (M.M.H.); Neuroscience Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts (B.L.K., P.R.V.);and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts (L.X.)
| | - Bethany L Kormos
- Neuroscience & Pain Research Unit, BioTherapeutics Research and Development, Pfizer Inc. Cambridge, Massachusetts (Q.W., S.P.A., D.M.R., N.J.B., M.D.E.); Center of Chemistry Innovation and Excellence, Pfizer Inc., Groton, Connecticut (M.M.H.); Neuroscience Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts (B.L.K., P.R.V.);and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts (L.X.)
| | - Patrick R Verhoest
- Neuroscience & Pain Research Unit, BioTherapeutics Research and Development, Pfizer Inc. Cambridge, Massachusetts (Q.W., S.P.A., D.M.R., N.J.B., M.D.E.); Center of Chemistry Innovation and Excellence, Pfizer Inc., Groton, Connecticut (M.M.H.); Neuroscience Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts (B.L.K., P.R.V.);and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts (L.X.)
| | - Lan Xu
- Neuroscience & Pain Research Unit, BioTherapeutics Research and Development, Pfizer Inc. Cambridge, Massachusetts (Q.W., S.P.A., D.M.R., N.J.B., M.D.E.); Center of Chemistry Innovation and Excellence, Pfizer Inc., Groton, Connecticut (M.M.H.); Neuroscience Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts (B.L.K., P.R.V.);and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts (L.X.)
| | - Nicholas J Brandon
- Neuroscience & Pain Research Unit, BioTherapeutics Research and Development, Pfizer Inc. Cambridge, Massachusetts (Q.W., S.P.A., D.M.R., N.J.B., M.D.E.); Center of Chemistry Innovation and Excellence, Pfizer Inc., Groton, Connecticut (M.M.H.); Neuroscience Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts (B.L.K., P.R.V.);and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts (L.X.)
| | - Michael D Ehlers
- Neuroscience & Pain Research Unit, BioTherapeutics Research and Development, Pfizer Inc. Cambridge, Massachusetts (Q.W., S.P.A., D.M.R., N.J.B., M.D.E.); Center of Chemistry Innovation and Excellence, Pfizer Inc., Groton, Connecticut (M.M.H.); Neuroscience Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts (B.L.K., P.R.V.);and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts (L.X.)
| |
Collapse
|
143
|
Bodea CA, Middleton FA, Melhem NM, Klei L, Song Y, Tiobech J, Marumoto P, Yano V, Faraone SV, Roeder K, Myles-Worsley M, Devlin B, Byerley W. Analysis of Shared Haplotypes amongst Palauans Maps Loci for Psychotic Disorders to 4q28 and 5q23-q31. Complex Psychiatry 2016; 2:173-184. [DOI: 10.1159/000450726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 08/19/2016] [Indexed: 11/19/2022] Open
Abstract
To localize genetic variation affecting risk for psychotic disorders in the population of Palau, we genotyped DNA samples from 203 Palauan individuals diagnosed with psychotic disorders, broadly defined, and 125 control subjects using a genome-wide single nucleotide polymorphism array. Palau has unique features advantageous for this study: due to its population history, Palauans are substantially interrelated; affected individuals often, but not always, cluster in families; and we have essentially complete ascertainment of affected individuals. To localize risk variants to genomic regions, we evaluated long-shared haplotypes, ≥10 Mb, identifying clusters of affected individuals who share such haplotypes. This extensive sharing, typically identical by descent, was significantly greater in cases than population controls, even after controlling for relatedness. Several regions of the genome exhibited substantial excess of shared haplotypes for affected individuals, including 3p21, 3p12, 4q28, and 5q23-q31. Two of these regions, 4q28 and 5q23-q31, showed significant linkage by traditional LOD score analysis and could harbor variants of more sizeable risk for psychosis or a multiplicity of risk variants. The pattern of haplotype sharing in 4q28 highlights <i>PCDH10</i>, encoding a cadherin-related neuronal receptor, as possibly involved in risk.
Collapse
|
144
|
Smoller JW. The Genetics of Stress-Related Disorders: PTSD, Depression, and Anxiety Disorders. Neuropsychopharmacology 2016; 41:297-319. [PMID: 26321314 PMCID: PMC4677147 DOI: 10.1038/npp.2015.266] [Citation(s) in RCA: 295] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/05/2015] [Accepted: 08/26/2015] [Indexed: 02/06/2023]
Abstract
Research into the causes of psychopathology has largely focused on two broad etiologic factors: genetic vulnerability and environmental stressors. An important role for familial/heritable factors in the etiology of a broad range of psychiatric disorders was established well before the modern era of genomic research. This review focuses on the genetic basis of three disorder categories-posttraumatic stress disorder (PTSD), major depressive disorder (MDD), and the anxiety disorders-for which environmental stressors and stress responses are understood to be central to pathogenesis. Each of these disorders aggregates in families and is moderately heritable. More recently, molecular genetic approaches, including genome-wide studies of genetic variation, have been applied to identify specific risk variants. In this review, I summarize evidence for genetic contributions to PTSD, MDD, and the anxiety disorders including genetic epidemiology, the role of common genetic variation, the role of rare and structural variation, and the role of gene-environment interaction. Available data suggest that stress-related disorders are highly complex and polygenic and, despite substantial progress in other areas of psychiatric genetics, few risk loci have been identified for these disorders. Progress in this area will likely require analysis of much larger sample sizes than have been reported to date. The phenotypic complexity and genetic overlap among these disorders present further challenges. The review concludes with a discussion of prospects for clinical translation of genetic findings and future directions for research.
Collapse
Affiliation(s)
- Jordan W Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
145
|
Monin A, Fournier M, Baumann PS, Cuénod M, Do KQ. Role of Redox Dysregulation in White Matter Anomalies Associated with Schizophrenia. HANDBOOK OF BEHAVIORAL NEUROSCIENCE 2016. [DOI: 10.1016/b978-0-12-800981-9.00028-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
146
|
Liu JR, Liu Q, Khoury J, Li YJ, Han XH, Li J, Ibla JC. Hypoxic preconditioning decreases nuclear factor κB activity via Disrupted in Schizophrenia-1. Int J Biochem Cell Biol 2016; 70:140-148. [PMID: 26615762 DOI: 10.1016/j.biocel.2015.11.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/19/2015] [Accepted: 11/20/2015] [Indexed: 01/15/2023]
Abstract
Nuclear factor κB is a key mediator of inflammation during conditions of hypoxia. Here, we used models of hypoxic pre-conditioning as mechanism to decrease nuclear factor κB activity induced by hypoxia. Our initial studies suggested that Disrupted in Schizophrenia-1 may be induced by hypoxic pre-conditioning and possibly involved in the regulation of nuclear factor κB. In this study we used Disrupted in Schizophrenia-1 exogenous over-expression and knock-down to determine its effect on ataxia telangiectasia mutated--nuclear factor κB activation cascade. Our results demonstrated that hypoxic pre-conditioning significantly increased the expression of Disrupted in Schizophrenia-1 at mRNA and protein levels both in vitro and in vivo. Over-expression of Disrupted in Schizophrenia-1 significantly attenuated the hypoxia-mediated ataxia telangiectasia mutated phosphorylation and prevented its cytoplasm translocation where it functions to activate nuclear factor κB. We further determined that Disrupted in Schizophrenia-1 activated the protein phosphatase 2A, preventing the phosphorylation of ataxia telangiectasia mutated serine-1981, the main regulatory site of ataxia telangiectasia mutated activity. Cellular levels of Disrupted in Schizophrenia-1 protein significantly decreased nuclear factor κB activation profiles and pro-inflammatory gene expression. Taken together, these results demonstrate that hypoxic pre-conditioning decreases the activation of nuclear factor κB through the transcriptional induction of Disrupted in Schizophrenia-1.
Collapse
Affiliation(s)
- Jia-Ren Liu
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, United States.
| | - Qian Liu
- Department of Pediatric Surgery, The First Affiliated Hospital of GanNan Medical University, JiangXi 341000, PR China
| | - Joseph Khoury
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, United States
| | - Yue-Jin Li
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, United States
| | - Xiao-Hui Han
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, United States
| | - Jing Li
- Laboratory of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, PR China
| | - Juan C Ibla
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, United States.
| |
Collapse
|
147
|
Abstract
Endophenotypes are quantitative, heritable traits that may help to elucidate the pathophysiologic mechanisms underlying complex disease syndromes, such as schizophrenia. They can be assessed at numerous levels of analysis; here, we review electrophysiological endophenotypes that have shown promise in helping us understand schizophrenia from a more mechanistic point of view. For each endophenotype, we describe typical experimental procedures, reliability, heritability, and reported gene and neurobiological associations. We discuss recent findings regarding the genetic architecture of specific electrophysiological endophenotypes, as well as converging evidence from EEG studies implicating disrupted balance of glutamatergic signaling and GABAergic inhibition in the pathophysiology of schizophrenia. We conclude that refining the measurement of electrophysiological endophenotypes, expanding genetic association studies, and integrating data sets are important next steps for understanding the mechanisms that connect identified genetic risk loci for schizophrenia to the disease phenotype.
Collapse
Affiliation(s)
- Emily Owens
- Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA
| | - Peter Bachman
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - David C Glahn
- Olin Neuropsychiatric Research Center, Institute of Living, Hartford, CT,Department of Psychiatry, Yale University School of Medicine, New Haven, CT
| | - Carrie E Bearden
- Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA
| |
Collapse
|
148
|
Caution When Diagnosing Your Mouse With Schizophrenia: The Use and Misuse of Model Animals for Understanding Psychiatric Disorders. Biol Psychiatry 2016; 79:32-8. [PMID: 26058706 DOI: 10.1016/j.biopsych.2015.04.023] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 03/26/2015] [Accepted: 04/23/2015] [Indexed: 12/14/2022]
Abstract
Animal models are widely used in biomedical research, but their applicability to psychiatric disorders is less clear. There are several reasons for this, including 1) emergent features of psychiatric illness that are not captured by the sum of individual symptoms, 2) a lack of equivalency between model animal behavior and human psychiatric symptoms, and 3) the possibility that model organisms do not have (and may not be capable of having) the same illnesses as humans. Here, we discuss the effective use, and inherent limitations, of model animals for psychiatric research. As disrupted-in-schizophrenia 1 (DISC1) is a genetic risk factor across a spectrum of psychiatric disorders, we focus on the results of studies using mice with various mutations of DISC1. The data from a broad range of studies show remarkable consistency with the effects of DISC1 mutation on developmental/anatomical endophenotypes. However, when one expands the phenotype to include behavioral correlates of human psychiatric diseases, much of this consistency ends. Despite these challenges, model animals remain valuable for understanding the basic brain processes that underlie psychiatric diseases. We argue that model animals have great potential to help us understand the core neurobiological dysfunction underlying psychiatric disorders and that marrying genetics and brain circuits with behavior is a good way forward.
Collapse
|
149
|
Greenwood TA, Lazzeroni LC, Calkins ME, Freedman R, Green MF, Gur RE, Gur RC, Light GA, Nuechterlein KH, Olincy A, Radant AD, Seidman LJ, Siever LJ, Silverman JM, Stone WS, Sugar CA, Swerdlow NR, Tsuang DW, Tsuang MT, Turetsky BI, Braff DL. Genetic assessment of additional endophenotypes from the Consortium on the Genetics of Schizophrenia Family Study. Schizophr Res 2016; 170:30-40. [PMID: 26597662 PMCID: PMC4707095 DOI: 10.1016/j.schres.2015.11.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 11/06/2015] [Accepted: 11/10/2015] [Indexed: 01/15/2023]
Abstract
The Consortium on the Genetics of Schizophrenia Family Study (COGS-1) has previously reported our efforts to characterize the genetic architecture of 12 primary endophenotypes for schizophrenia. We now report the characterization of 13 additional measures derived from the same endophenotype test paradigms in the COGS-1 families. Nine of the measures were found to discriminate between schizophrenia patients and controls, were significantly heritable (31 to 62%), and were sufficiently independent of previously assessed endophenotypes, demonstrating utility as additional endophenotypes. Genotyping via a custom array of 1536 SNPs from 94 candidate genes identified associations for CTNNA2, ERBB4, GRID1, GRID2, GRIK3, GRIK4, GRIN2B, NOS1AP, NRG1, and RELN across multiple endophenotypes. An experiment-wide p value of 0.003 suggested that the associations across all SNPs and endophenotypes collectively exceeded chance. Linkage analyses performed using a genome-wide SNP array further identified significant or suggestive linkage for six of the candidate endophenotypes, with several genes of interest located beneath the linkage peaks (e.g., CSMD1, DISC1, DLGAP2, GRIK2, GRIN3A, and SLC6A3). While the partial convergence of the association and linkage likely reflects differences in density of gene coverage provided by the distinct genotyping platforms, it is also likely an indication of the differential contribution of rare and common variants for some genes and methodological differences in detection ability. Still, many of the genes implicated by COGS through endophenotypes have been identified by independent studies of common, rare, and de novo variation in schizophrenia, all converging on a functional genetic network related to glutamatergic neurotransmission that warrants further investigation.
Collapse
Affiliation(s)
- Tiffany A Greenwood
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States.
| | - Laura C Lazzeroni
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
| | - Monica E Calkins
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, United States
| | - Robert Freedman
- Department of Psychiatry, University of Colorado Health Sciences Center, Denver, CO, United States
| | - Michael F Green
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, United States; VA Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Raquel E Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, United States
| | - Ruben C Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, United States
| | - Gregory A Light
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States; VISN-22 Mental Illness, Research, Education and Clinical Center (MIRECC), VA San Diego Healthcare System, United States
| | - Keith H Nuechterlein
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, United States
| | - Ann Olincy
- Department of Psychiatry, University of Colorado Health Sciences Center, Denver, CO, United States
| | - Allen D Radant
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, United States; VA Puget Sound Health Care System, Seattle, WA, United States
| | - Larry J Seidman
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States; Massachusetts Mental Health Center Public Psychiatry Division of the Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Larry J Siever
- Department of Psychiatry, The Mount Sinai School of Medicine, New York, NY, United States; James J. Peters VA Medical Center, New York, NY, United States
| | - Jeremy M Silverman
- Department of Psychiatry, The Mount Sinai School of Medicine, New York, NY, United States; James J. Peters VA Medical Center, New York, NY, United States
| | - William S Stone
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States; Massachusetts Mental Health Center Public Psychiatry Division of the Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Catherine A Sugar
- Department of Biostatistics, University of California Los Angeles School of Public Health, Los Angeles, CA, United States
| | - Neal R Swerdlow
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | - Debby W Tsuang
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, United States; VA Puget Sound Health Care System, Seattle, WA, United States
| | - Ming T Tsuang
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States; Center for Behavioral Genomics, Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, United States; Harvard Institute of Psychiatric Epidemiology and Genetics, Boston, MA, United States
| | - Bruce I Turetsky
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, United States
| | - David L Braff
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States; VISN-22 Mental Illness, Research, Education and Clinical Center (MIRECC), VA San Diego Healthcare System, United States
| |
Collapse
|
150
|
Abstract
Recent technological advances in next-generation sequencing (NGS) provide unprecedented power to sequence personal genomes, characterize genomic landscapes, and detect a large number of sequence variants. The discovery of disease-causing variants in patients' genomes has dramatically changed our perspective on precision medicine. This chapter provides an overview of sequence variant detection and analysis in NGS study. We outline the general methods for identifying different types of sequence variants from NGS data. We summarize the common approaches for analyzing and visualizing casual variants associated with complex diseases on precision medicine informatics.
Collapse
Affiliation(s)
- Shaolei Teng
- Department of Biology, Howard University, Washington, DC, 20059, USA.
| |
Collapse
|