1
|
Shokrgozar A, Rahimi M, Shoraka S. Identification of key genes and pathways in schizophrenia: a bioinformatics analysis based on GWAS and GEO. Front Psychiatry 2025; 16:1464676. [PMID: 40259965 PMCID: PMC12009944 DOI: 10.3389/fpsyt.2025.1464676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 03/14/2025] [Indexed: 04/23/2025] Open
Abstract
Introduction Schizophrenia is a mental illness that is associated with many disorders, such as incoherence of mental activities, and impairment of perception, thinking, emotions, and behavior. Although the exact cause of schizophrenia is unknown, many studies have highlighted the role of genetic background and environmental factors in this disease. Therefore, the identification of key genes involved in schizophrenia provides a promising opportunity to develop novel diagnosis and/or treatment methods. This study aims to investigate schizophrenia-related hub genes by bioinformatics analysis based on genome-wide association (GWAS) and gene expression omnibus (GEO) datasets. Material and methods In this study, the GWAS catalog and GEO dataset were used to identify key candidate genes and pathways that are important in the diagnosis and treatment of schizophrenia, and then the results were analyzed using Enrichr and Cytoscape tools. Result According to our result NRXN, CACNA1C, and GRIN2A genes had the highest scores in the GWAS analyses and BRCA1, ATM, and STAT1 genes had the highest scores in the GEO dataset. Also, glucuronidation, ascorbate, and aldarate metabolism pathways in the GWAS, PI3K/AKT and Rap1 signaling in the GEO had the highest associations with schizophrenia. Conclusion This study highlights the need for further validation of the genes and molecular pathways in schizophrenia. Also, the identified genes could be promising candidates for future diagnostic and/or treatment strategies for schizophrenia.
Collapse
Affiliation(s)
| | - Maryam Rahimi
- Clinical Care and Health Promotion Research Center, Islamic Azad University, Karaj, Iran
| | - Shahrzad Shoraka
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
2
|
Khalil M, Kalyoncu A, Bellon A. Genetics of Suicide. Genes (Basel) 2025; 16:428. [PMID: 40282388 PMCID: PMC12027201 DOI: 10.3390/genes16040428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Over the past two decades, suicide has consistently ranked among the leading causes of death in the United States. While suicide deaths are closely associated with uicidal ideation and attempts, these are not good predictors of future suicide deaths. Establishing who is at risk of suicide remains a challenge that is mostly hampered by the lack of understanding of its pathophysiology. Nonetheless, evidence continues to accumulate suggesting that suicide is driven by a complex and dynamic interaction between environmental factors and genetics. The identification of genes that place people at risk of suicide remains elusive, but data are rapidly evolving. In this narrative review, we describe how Tryptophan hydroxylase (TPH) genes, particularly TPH1 and TPH2, have been associated with suicide in various publications. There is also replicated evidence linking the brain-derived neurotrophic factor gene to suicide, with its most consistent results originating from epigenetic studies. Not surprisingly, many genes involved in the hypothalamic-pituitary-adrenal axis have been connected with suicide, but these data require replication. Finally, among the inflammatory genes studied in suicide, only specific polymorphisms in TNF-alpha and IL-6 may increase susceptibility to suicidal behavior. In conclusion, significant work remains to be performed as inconsistencies undermine the reliability of genetic results in suicide. Potential avenues for future research are proposed.
Collapse
Affiliation(s)
- Mostafa Khalil
- Brown University, Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Providence, RI 02912, USA;
| | - Anil Kalyoncu
- Penn State Hershey Medical Center, Department of Psychiatry and Behavioral Health, Hershey, PA 17033, USA;
| | - Alfredo Bellon
- Penn State Hershey Medical Center, Department of Psychiatry and Behavioral Health, Hershey, PA 17033, USA;
- Penn State Hershey Medical Center, Department of Pharmacology, Hershey, PA 17033, USA
| |
Collapse
|
3
|
Hatzimanolis O, Sykes AM, Cristino AS. Circular RNAs in neurological conditions - computational identification, functional validation, and potential clinical applications. Mol Psychiatry 2025; 30:1652-1675. [PMID: 39966624 PMCID: PMC11919710 DOI: 10.1038/s41380-025-02925-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 01/11/2025] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
Non-coding RNAs (ncRNAs) have gained significant attention in recent years due to advancements in biotechnology, particularly high-throughput total RNA sequencing. These developments have led to new understandings of non-coding biology, revealing that approximately 80% of non-coding regions in the genome possesses biochemical functionality. Among ncRNAs, circular RNAs (circRNAs), first identified in 1976, have emerged as a prominent research field. CircRNAs are abundant in most human cell types, evolutionary conserved, highly stable, and formed by back-splicing events which generate covalently closed ends. Notably, circRNAs exhibit high expression levels in neural tissue and perform diverse biochemical functions, including acting as molecular sponges for microRNAs, interacting with RNA-binding proteins to regulate their availability and activity, modulating transcription and splicing, and even translating into functional peptides in some cases. Recent advancements in computational and experimental methods have enhanced our ability to identify and validate circRNAs, providing valuable insights into their biological roles. This review focuses on recent developments in circRNA research as they related to neuropsychiatric and neurodegenerative conditions. We also explore their potential applications in clinical diagnostics, therapeutics, and future research directions. CircRNAs remain a relatively underexplored area of non-coding biology, particularly in the context of neurological disorders. However, emerging evidence supports their role as critical players in the etiology and molecular mechanisms of conditions such as schizophrenia, bipolar disorder, major depressive disorder, Alzheimer's disease, and Parkinson's disease. These findings suggest that circRNAs may provide a novel framework contributing to the molecular dysfunctions underpinning these complex neurological conditions.
Collapse
Affiliation(s)
- Oak Hatzimanolis
- Institute for Biomedicine and Glycomics, Griffith University, Brisbane, QLD, Australia
| | - Alex M Sykes
- Institute for Biomedicine and Glycomics, Griffith University, Brisbane, QLD, Australia
| | - Alexandre S Cristino
- Institute for Biomedicine and Glycomics, Griffith University, Brisbane, QLD, Australia.
| |
Collapse
|
4
|
Montalbano S, Krebs MD, Rosengren A, Vaez M, Hellberg KLG, Mortensen PB, Børglum AD, Geschwind DH, Raznahan A, Thompson WK, Helenius D, Werge T, Ingason A. Analysis of exonic deletions in a large population study provides novel insights into NRXN1 pathology. NPJ Genom Med 2024; 9:67. [PMID: 39695155 DOI: 10.1038/s41525-024-00450-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024] Open
Abstract
The NRXN1 locus is a hotspot for non-recurrent copy number variants and exon-disrupting NRXN1 deletions have been associated with increased risk of neurodevelopmental disorders in case-control studies. However, corresponding population-based estimates of prevalence and disease-associated risk are currently lacking. Also, most studies have not differentiated between deletions affecting exons of different NRXN1 splice variants nor considered intronic deletions. We used the iPSYCH2015 case-cohort sample to obtain unbiased estimates of the prevalence of NRXN1 deletions and their associated risk of autism, schizophrenia, depression, and ADHD. Most exon-disrupting deletions affected exons specific to the alpha isoform, and almost half of the non-exonic deletions represented a previously reported segregating founder deletion. Carriage of exon-disrupting NRXN1 deletions was associated with a threefold and twofold increased risk of autism and ADHD, respectively, whereas no significantly increased risk of depression or schizophrenia was observed. Our results highlight the importance of using population-based samples in genetic association studies.
Collapse
Affiliation(s)
- Simone Montalbano
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Roskilde, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen and Aarhus, Denmark
| | - Morten Dybdahl Krebs
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Roskilde, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen and Aarhus, Denmark
| | - Anders Rosengren
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Roskilde, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen and Aarhus, Denmark
| | - Morteza Vaez
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Roskilde, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen and Aarhus, Denmark
| | - Kajsa-Lotta Georgii Hellberg
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Roskilde, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen and Aarhus, Denmark
| | - Preben B Mortensen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen and Aarhus, Denmark
- National Centre for Register-based Research, Aarhus University, Aarhus, Denmark
| | - Anders D Børglum
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen and Aarhus, Denmark
- Department of Biomedicine - Human Genetics and the iSEQ Center, Aarhus University, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, Aarhus, Denmark
| | - Daniel H Geschwind
- Department of Neurology, University of California, Los Angeles, CA, USA
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Armin Raznahan
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health Intramural Research Program, Bethesda, MD, USA
| | - Wesley K Thompson
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen and Aarhus, Denmark
- Laureate Institute for Brain Research, Tulsa, OK, USA
| | - Dorte Helenius
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Roskilde, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen and Aarhus, Denmark
| | - Thomas Werge
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Roskilde, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen and Aarhus, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Andrés Ingason
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Roskilde, Denmark.
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen and Aarhus, Denmark.
| |
Collapse
|
5
|
Mohrmann L, Seebach J, Missler M, Rohlmann A. Distinct Alterations in Dendritic Spine Morphology in the Absence of β-Neurexins. Int J Mol Sci 2024; 25:1285. [PMID: 38279285 PMCID: PMC10817056 DOI: 10.3390/ijms25021285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/12/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
Dendritic spines are essential for synaptic function because they constitute the postsynaptic compartment of the neurons that receives the most excitatory input. The extracellularly shorter variant of the presynaptic cell adhesion molecules neurexins, β-neurexin, has been implicated in various aspects of synaptic function, including neurotransmitter release. However, its role in developing or stabilizing dendritic spines as fundamental computational units of excitatory synapses has remained unclear. Here, we show through morphological analysis that the deletion of β-neurexins in hippocampal neurons in vitro and in hippocampal tissue in vivo affects presynaptic dense-core vesicles, as hypothesized earlier, and, unexpectedly, alters the postsynaptic spine structure. Specifically, we observed that the absence of β-neurexins led to an increase in filopodial-like protrusions in vitro and more mature mushroom-type spines in the CA1 region of adult knockout mice. In addition, the deletion of β-neurexins caused alterations in the spine head dimension and an increase in spines with perforations of their postsynaptic density but no changes in the overall number of spines or synapses. Our results indicate that presynaptic β-neurexins play a role across the synaptic cleft, possibly by aligning with postsynaptic binding partners and glutamate receptors via transsynaptic columns.
Collapse
Affiliation(s)
| | | | - Markus Missler
- Institute of Anatomy and Molecular Neurobiology, University Münster, 48149 Münster, Germany; (L.M.); (J.S.)
| | - Astrid Rohlmann
- Institute of Anatomy and Molecular Neurobiology, University Münster, 48149 Münster, Germany; (L.M.); (J.S.)
| |
Collapse
|
6
|
Maury EA, Sherman MA, Genovese G, Gilgenast TG, Kamath T, Burris S, Rajarajan P, Flaherty E, Akbarian S, Chess A, McCarroll SA, Loh PR, Phillips-Cremins JE, Brennand KJ, Macosko EZ, Walters JT, O’Donovan M, Sullivan P, Sebat J, Lee EA, Walsh CA. Schizophrenia-associated somatic copy-number variants from 12,834 cases reveal recurrent NRXN1 and ABCB11 disruptions. CELL GENOMICS 2023; 3:100356. [PMID: 37601975 PMCID: PMC10435376 DOI: 10.1016/j.xgen.2023.100356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/21/2022] [Accepted: 06/09/2023] [Indexed: 08/22/2023]
Abstract
While germline copy-number variants (CNVs) contribute to schizophrenia (SCZ) risk, the contribution of somatic CNVs (sCNVs)-present in some but not all cells-remains unknown. We identified sCNVs using blood-derived genotype arrays from 12,834 SCZ cases and 11,648 controls, filtering sCNVs at loci recurrently mutated in clonal blood disorders. Likely early-developmental sCNVs were more common in cases (0.91%) than controls (0.51%, p = 2.68e-4), with recurrent somatic deletions of exons 1-5 of the NRXN1 gene in five SCZ cases. Hi-C maps revealed ectopic, allele-specific loops forming between a potential cryptic promoter and non-coding cis-regulatory elements upon 5' deletions in NRXN1. We also observed recurrent intragenic deletions of ABCB11, encoding a transporter implicated in anti-psychotic response, in five treatment-resistant SCZ cases and showed that ABCB11 is specifically enriched in neurons forming mesocortical and mesolimbic dopaminergic projections. Our results indicate potential roles of sCNVs in SCZ risk.
Collapse
Affiliation(s)
- Eduardo A. Maury
- Division of Genetics and Genomics, Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Bioinformatics & Integrative Genomics Program and Harvard/MIT MD-PHD Program, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Maxwell A. Sherman
- Brigham and Women’s Hospital, Division of Genetics & Center for Data Sciences, Boston, MA, USA
| | - Giulio Genovese
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Thomas G. Gilgenast
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Tushar Kamath
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Graduate Program in Biophysics, Harvard University, Cambridge, MA, USA
| | - S.J. Burris
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Prashanth Rajarajan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Department of Genetics & Genomics, Icahn Institute of Genomics and Multiscale Biology, Department of Psychiatry, Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine of Mount Sinai, New York, NY, USA
| | - Erin Flaherty
- Nash Family Department of Neuroscience, Friedman Brain Institute, Department of Genetics & Genomics, Icahn Institute of Genomics and Multiscale Biology, Department of Psychiatry, Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine of Mount Sinai, New York, NY, USA
| | - Schahram Akbarian
- Nash Family Department of Neuroscience, Friedman Brain Institute, Department of Genetics & Genomics, Icahn Institute of Genomics and Multiscale Biology, Department of Psychiatry, Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine of Mount Sinai, New York, NY, USA
| | - Andrew Chess
- Nash Family Department of Neuroscience, Friedman Brain Institute, Department of Genetics & Genomics, Icahn Institute of Genomics and Multiscale Biology, Department of Psychiatry, Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine of Mount Sinai, New York, NY, USA
| | - Steven A. McCarroll
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Po-Ru Loh
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital, Division of Genetics & Center for Data Sciences, Boston, MA, USA
| | | | - Kristen J. Brennand
- Nash Family Department of Neuroscience, Friedman Brain Institute, Department of Genetics & Genomics, Icahn Institute of Genomics and Multiscale Biology, Department of Psychiatry, Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine of Mount Sinai, New York, NY, USA
- Departments of Psychiatry and Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Evan Z. Macosko
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital, Department of Psychiatry, Boston, MA, USA
| | - James T.R. Walters
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychiatry and Clinical Neurosciences, Cardiff University, Cardiff, Wales
| | - Michael O’Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychiatry and Clinical Neurosciences, Cardiff University, Cardiff, Wales
| | - Patrick Sullivan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jonathan Sebat
- University of California San Diego, Department of Psychiatry, Department of Cellular & Molecular Medicine, Beyster Center of Psychiatric Genomics, San Diego, CA, USA
| | - Eunjung A. Lee
- Division of Genetics and Genomics, Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Christopher A. Walsh
- Division of Genetics and Genomics, Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA, USA
| |
Collapse
|
7
|
Farsi Z, Sheng M. Molecular mechanisms of schizophrenia: Insights from human genetics. Curr Opin Neurobiol 2023; 81:102731. [PMID: 37245257 DOI: 10.1016/j.conb.2023.102731] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/28/2023] [Accepted: 05/01/2023] [Indexed: 05/30/2023]
Abstract
Schizophrenia is a debilitating psychiatric disorder that affects millions of people worldwide; however, its etiology is poorly understood at the molecular and neurobiological levels. A particularly important advance in recent years is the discovery of rare genetic variants associated with a greatly increased risk of developing schizophrenia. These primarily loss-of-function variants are found in genes that overlap with those implicated by common variants and are involved in the regulation of glutamate signaling, synaptic function, DNA transcription, and chromatin remodeling. Animal models harboring mutations in these large-effect schizophrenia risk genes show promise in providing additional insights into the molecular mechanisms of the disease.
Collapse
Affiliation(s)
- Zohreh Farsi
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Morgan Sheng
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
8
|
Lin JR, Zhao Y, Jabalameli MR, Nguyen N, Mitra J, Swillen A, Vorstman JAS, Chow EWC, van den Bree M, Emanuel BS, Vermeesch JR, Owen MJ, Williams NM, Bassett AS, McDonald-McGinn DM, Gur RE, Bearden CE, Morrow BE, Lachman HM, Zhang ZD. Rare coding variants as risk modifiers of the 22q11.2 deletion implicate postnatal cortical development in syndromic schizophrenia. Mol Psychiatry 2023; 28:2071-2080. [PMID: 36869225 PMCID: PMC11793908 DOI: 10.1038/s41380-023-02009-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/15/2023] [Accepted: 02/21/2023] [Indexed: 03/05/2023]
Abstract
22q11.2 deletion is one of the strongest known genetic risk factors for schizophrenia. Recent whole-genome sequencing of schizophrenia cases and controls with this deletion provided an unprecedented opportunity to identify risk modifying genetic variants and investigate their contribution to the pathogenesis of schizophrenia in 22q11.2 deletion syndrome. Here, we apply a novel analytic framework that integrates gene network and phenotype data to investigate the aggregate effects of rare coding variants and identified modifier genes in this etiologically homogenous cohort (223 schizophrenia cases and 233 controls of European descent). Our analyses revealed significant additive genetic components of rare nonsynonymous variants in 110 modifier genes (adjusted P = 9.4E-04) that overall accounted for 4.6% of the variance in schizophrenia status in this cohort, of which 4.0% was independent of the common polygenic risk for schizophrenia. The modifier genes affected by rare coding variants were enriched with genes involved in synaptic function and developmental disorders. Spatiotemporal transcriptomic analyses identified an enrichment of coexpression between modifier and 22q11.2 genes in cortical brain regions from late infancy to young adulthood. Corresponding gene coexpression modules are enriched with brain-specific protein-protein interactions of SLC25A1, COMT, and PI4KA in the 22q11.2 deletion region. Overall, our study highlights the contribution of rare coding variants to the SCZ risk. They not only complement common variants in disease genetics but also pinpoint brain regions and developmental stages critical to the etiology of syndromic schizophrenia.
Collapse
Affiliation(s)
- Jhih-Rong Lin
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA
| | - Yingjie Zhao
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA
| | - M Reza Jabalameli
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA
| | - Nha Nguyen
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA
| | - Joydeep Mitra
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA
| | - Ann Swillen
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | | | - Eva W C Chow
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Marianne van den Bree
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Beverly S Emanuel
- Division of Human Genetics and 22q and You Center, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Michael J Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Nigel M Williams
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Anne S Bassett
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Donna M McDonald-McGinn
- Division of Human Genetics and 22q and You Center, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Raquel E Gur
- Department of Psychiatry and Lifespan Brain Institute, Penn Medicine-CHOP, University of Pennsylvania, Philadelphia, PA, USA
| | - Carrie E Bearden
- Departments of Psychiatry and Biobehavioral Sciences and Psychology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Bernice E Morrow
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA
| | - Herbert M Lachman
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA
| | - Zhengdong D Zhang
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA.
| |
Collapse
|
9
|
Wang H, Wang LS, Schellenberg G, Lee WP. The role of structural variations in Alzheimer's disease and other neurodegenerative diseases. Front Aging Neurosci 2023; 14:1073905. [PMID: 36846102 PMCID: PMC9944073 DOI: 10.3389/fnagi.2022.1073905] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/31/2022] [Indexed: 02/10/2023] Open
Abstract
Dozens of single nucleotide polymorphisms (SNPs) related to Alzheimer's disease (AD) have been discovered by large scale genome-wide association studies (GWASs). However, only a small portion of the genetic component of AD can be explained by SNPs observed from GWAS. Structural variation (SV) can be a major contributor to the missing heritability of AD; while SV in AD remains largely unexplored as the accurate detection of SVs from the widely used array-based and short-read technology are still far from perfect. Here, we briefly summarized the strengths and weaknesses of available SV detection methods. We reviewed the current landscape of SV analysis in AD and SVs that have been found associated with AD. Particularly, the importance of currently less explored SVs, including insertions, inversions, short tandem repeats, and transposable elements in neurodegenerative diseases were highlighted.
Collapse
Affiliation(s)
- Hui Wang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Li-San Wang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Gerard Schellenberg
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Wan-Ping Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
10
|
Lee CC, Ye R, Tubbs JD, Baum L, Zhong Y, Leung SYJ, Chan SC, Wu KYK, Cheng PKJ, Chow LP, Leung PWL, Sham PC. Third-generation genome sequencing implicates medium-sized structural variants in chronic schizophrenia. Front Neurosci 2023; 16:1058359. [PMID: 36711134 PMCID: PMC9874699 DOI: 10.3389/fnins.2022.1058359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/14/2022] [Indexed: 01/13/2023] Open
Abstract
Background Schizophrenia (SCZ) is a heterogeneous psychiatric disorder, with significant contribution from genetic factors particularly for chronic cases with negative symptoms and cognitive deficits. To date, Genome Wide Association Studies (GWAS) and exome sequencing have associated SCZ with a number of single nucleotide polymorphisms (SNPs) and copy number variants (CNVs), but there is still missing heritability. Medium-sized structural variants (SVs) are difficult to detect using SNP arrays or second generation sequencing, and may account for part of the missing heritability of SCZ. Aims and objectives To identify SVs associated with severe chronic SCZ across the whole genome. Study design 10 multiplex families with probands suffering from chronic SCZ with negative symptoms and cognitive deficits were recruited, with all their affected members demonstrating uni-lineal inheritance. Control subjects comprised one affected member from the affected lineage, and unaffected members from each paternal and maternal lineage. Methods Third generation sequencing was applied to peripheral blood samples from 10 probands and 5 unaffected controls. Bioinformatic tools were used to identify SVs from the long sequencing reads, with confirmation of findings in probands by short-read Illumina sequencing, Sanger sequencing and visual manual validation with Integrated Genome Browser. Results In the 10 probands, we identified and validated 88 SVs (mostly in introns and medium-sized), within 79 genes, which were absent in the 5 unaffected control subjects. These 79 genes were enriched in 20 biological pathways which were related to brain development, neuronal migration, neurogenesis, neuronal/synaptic function, learning/memory, and hearing. These identified SVs also showed evidence for enrichment of genes that are highly expressed in the adolescent striatum. Conclusion A substantial part of the missing heritability in SCZ may be explained by medium-sized SVs detectable only by third generation sequencing. We have identified a number of such SVs potentially conferring risk for SCZ, which implicate multiple brain-related genes and pathways. In addition to previously-identified pathways involved in SCZ such as neurodevelopment and neuronal/synaptic functioning, we also found novel evidence for enrichment in hearing-related pathways and genes expressed in the adolescent striatum.
Collapse
Affiliation(s)
- Chi Chiu Lee
- Department of Psychiatry, Kwai Chung Hospital, Hong Kong, Hong Kong SAR, China,*Correspondence: Chi Chiu Lee,
| | - Rui Ye
- Department of Psychiatry, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Justin D. Tubbs
- Department of Psychiatry, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Larry Baum
- Department of Psychiatry, The University of Hong Kong, Hong Kong, Hong Kong SAR, China,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yuanxin Zhong
- Department of Psychiatry, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Shuk Yan Joey Leung
- Department of Psychiatry, Kwai Chung Hospital, Hong Kong, Hong Kong SAR, China
| | - Sheung Chun Chan
- Department of Psychiatry, Tai Po Hospital, Hong Kong, Hong Kong SAR, China
| | - Kit Ying Kitty Wu
- Kowloon West Cluster, Hospital Authority, Hong Kong, Hong Kong SAR, China
| | - Po Kwan Jamie Cheng
- Department of Clinical Psychology, Yan Chai Hospital, Hong Kong, Hong Kong SAR, China
| | - Lai Ping Chow
- Department of Psychiatry, Kwai Chung Hospital, Hong Kong, Hong Kong SAR, China
| | - Patrick W. L. Leung
- Department of Psychology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Pak Chung Sham
- Department of Psychiatry, The University of Hong Kong, Hong Kong, Hong Kong SAR, China,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China,Centre for PanorOmic Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China,Pak Chung Sham,
| |
Collapse
|
11
|
Keshri N, Nandeesha H. Dysregulation of Synaptic Plasticity Markers in Schizophrenia. Indian J Clin Biochem 2023; 38:4-12. [PMID: 36684500 PMCID: PMC9852406 DOI: 10.1007/s12291-022-01068-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/05/2022] [Indexed: 01/25/2023]
Abstract
Schizophrenia is a mental disorder characterized by cognitive impairment resulting in compromised quality of life. Since the regulation of synaptic plasticity has functional implications in various aspects of cognition such as learning, memory, and neural circuit maturation, the dysregulation of synaptic plasticity is considered as a pathobiological feature of schizophrenia. The findings from our recently concluded studies indicate that there is an alteration in levels of synaptic plasticity markers such as neural cell adhesion molecule-1 (NCAM-1), Neurotropin-3 (NT-3) and Matrix-mettaloproteinase-9 (MMP-9) in schizophrenia patients. The objective of the present article is to review the role of markers of synaptic plasticity in schizophrenia. PubMed database (http;//www.ncbi.nlm.nih.gov/pubmed) was used to perform an extensive literature search using the keywords schizophrenia and synaptic plasticity. We conclude that markers of synaptic plasticity are altered in schizophrenia and may lead to complications of schizophrenia including cognitive dysfunction.
Collapse
Affiliation(s)
- Neha Keshri
- Department of Biochemistry, JIPMER, Puducherry, 605006 India
| | | |
Collapse
|
12
|
Arnsten AFT, Woo E, Yang S, Wang M, Datta D. Unusual Molecular Regulation of Dorsolateral Prefrontal Cortex Layer III Synapses Increases Vulnerability to Genetic and Environmental Insults in Schizophrenia. Biol Psychiatry 2022; 92:480-490. [PMID: 35305820 PMCID: PMC9372235 DOI: 10.1016/j.biopsych.2022.02.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/03/2022] [Accepted: 02/06/2022] [Indexed: 02/06/2023]
Abstract
Schizophrenia is associated with reduced numbers of spines and dendrites from layer III of the dorsolateral prefrontal cortex (dlPFC), the layer that houses the recurrent excitatory microcircuits that subserve working memory and abstract thought. Why are these synapses so vulnerable, while synapses in deeper or more superficial layers are little affected? This review describes the special molecular properties that govern layer III neurotransmission and neuromodulation in the primate dlPFC and how they may render these circuits particularly vulnerable to genetic and environmental insults. These properties include a reliance on NMDA receptor rather than AMPA receptor neurotransmission; cAMP (cyclic adenosine monophosphate) magnification of calcium signaling near the glutamatergic synapse of dendritic spines; and potassium channels opened by cAMP/PKA (protein kinase A) signaling that dynamically alter network strength, with built-in mechanisms to take dlPFC "offline" during stress. A variety of genetic and/or environmental insults can lead to the same phenotype of weakened layer III connectivity, in which mechanisms that normally strengthen connectivity are impaired and those that normally weaken connectivity are intensified. Inflammatory mechanisms, such as increased kynurenic acid and glutamate carboxypeptidase II expression, are especially detrimental to layer III dlPFC neurotransmission and modulation, mimicking genetic insults. The combination of genetic and inflammatory insults may cross the threshold into pathology.
Collapse
Affiliation(s)
- Amy F T Arnsten
- Department of Neuroscience, Yale Medical School, New Haven, Connecticut.
| | - Elizabeth Woo
- Department of Neuroscience, Yale Medical School, New Haven, Connecticut
| | - Shengtao Yang
- Department of Neuroscience, Yale Medical School, New Haven, Connecticut
| | - Min Wang
- Department of Neuroscience, Yale Medical School, New Haven, Connecticut
| | - Dibyadeep Datta
- Department of Neuroscience, Yale Medical School, New Haven, Connecticut
| |
Collapse
|
13
|
Kight KE, Argue KJ, Bumgardner JG, Bardhi K, Waddell J, McCarthy MM. Social behavior in prepubertal neurexin 1α deficient rats: A model of neurodevelopmental disorders. Behav Neurosci 2021; 135:782-803. [PMID: 34323517 PMCID: PMC8649076 DOI: 10.1037/bne0000482] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Loss-of-function mutations in the synaptic protein neurexin1α (NRXN1α) are associated with several neurodevelopmental disorders, including autism spectrum disorder (ASD), schizophrenia, and attention-deficit hyperactivity disorder (ADHD), and many of these disorders are defined by core deficits in social cognition. Mouse models of Nrxn1α deficiency are not amenable to studying aspects of social cognition because, in general, mice do not engage in complex social interactions such as social play or prosocial helping behaviors. Rats, on the contrary, engage in these complex, well-characterized social behaviors. Using the Nrxn1tm1Sage Sprague Dawley rat, we tested a range of cognitive and social behaviors in juveniles with haplo- or biallelic Nrxn1α mutation. We found a deficit in ultrasonic vocalizations (USVs) of male and female neonatal rats with Nrxn1α deficiency. A male-specific deficit in social play was observed in Nrxn1α-deficient juveniles, although sociability and social discrimination were unaltered. Nurturing behavior induced by exposure to pups was enhanced in male and female juveniles with biallelic Nrxn1α mutation. Performance in tasks of prosocial helping behavior and food retrieval indicated severe deficits in learning and cognition in juveniles with biallelic Nrxn1α mutation, and a less severe deficit in haploinsufficient rats, although Pavlovian learning was altered only in haploinsufficient males. We also observed a male-specific increase in mobility and object investigation in juveniles with complete Nrxn1α deficiency. Together, these observations more fully characterize the Nrxn1tm1Sage Sprague Dawley rat as a model for Nrxn1α-related neurodevelopmental disorders, and support a rationale for the juvenile rat as a more appropriate model for disorders that involve core deficits in complex social behaviors. (PsycInfo Database Record (c) 2021 APA, all rights reserved).
Collapse
Affiliation(s)
- Katherine E Kight
- Department of Pharmacology, University of Maryland School of Medicine
| | - Kathryn J Argue
- Department of Pharmacology, University of Maryland School of Medicine
| | | | - Keti Bardhi
- Department of Pediatrics, University of Maryland School of Medicine
| | - Jaylyn Waddell
- Department of Pediatrics, University of Maryland School of Medicine
| | | |
Collapse
|
14
|
Li Y, Ma C, Li W, Yang Y, Li X, Liu J, Wang J, Li S, Liu Y, Li K, Li J, Huang D, Chen R, Lv L, Li M, Luo XJ. A missense variant in NDUFA6 confers schizophrenia risk by affecting YY1 binding and NAGA expression. Mol Psychiatry 2021; 26:6896-6911. [PMID: 33931730 DOI: 10.1038/s41380-021-01125-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/31/2021] [Accepted: 04/13/2021] [Indexed: 12/18/2022]
Abstract
Genome-wide association studies (GWASs) have revealed that genetic variants at the 22q13.2 risk locus were robustly associated with schizophrenia. However, the causal variants at this risk locus and their roles in schizophrenia remain elusive. Here we identify the risk missense variant rs1801311 (located in the 1st exon of NDUFA6 gene) as likely causal for schizophrenia at 22q13.2 by disrupting binding of YY1, TAF1, and POLR2A. We systematically elucidated the regulatory mechanisms of rs1801311 and validated the regulatory effect of this missense variant. Intriguingly, rs1801311 physically interacted with NAGA (encodes the alpha-N-acetylgalactosaminidase, which is mainly involved in regulating metabolisms of glycoproteins and glycolipids in lysosome) and showed the most significant association with NAGA expression in the human brain, with the risk allele (G) associated with higher NAGA expression. Consistent with eQTL analysis, expression analysis showed that NAGA was significantly upregulated in brains of schizophrenia cases compared with controls, further supporting that rs1801311 may confer schizophrenia risk by regulating NAGA expression. Of note, we found that NAGA regulates important neurodevelopmental processes, including proliferation and differentiation of neural stem cells. Transcriptome analysis corroborated that NAGA regulates pathways associated with neuronal differentiation. Finally, we independently confirmed the association between rs1801311 and schizophrenia in a large Chinese cohort. Our study elucidates the regulatory mechanisms of the missense schizophrenia risk variant rs1801311 and provides mechanistic links between risk variant and schizophrenia etiology. In addition, this study also revealed the novel role of coding variants in gene regulation and schizophrenia risk, i.e., genetic variant in coding region of a specific gene may confer disease risk through regulating distal genes (act as regulatory variant for distal genes).
Collapse
Affiliation(s)
- Yifan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Changguo Ma
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Wenqiang Li
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China.,Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yongfeng Yang
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China.,Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xiaoyan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jiewei Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Junyang Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Shiwu Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yixing Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Kaiqin Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jiao Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Di Huang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Rui Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Luxian Lv
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China.,Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, Henan, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiong-Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China. .,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China. .,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan, China. .,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| |
Collapse
|
15
|
Priol AC, Denis L, Boulanger G, Thépaut M, Geoffray MM, Tordjman S. Detection of Morphological Abnormalities in Schizophrenia: An Important Step to Identify Associated Genetic Disorders or Etiologic Subtypes. Int J Mol Sci 2021; 22:ijms22179464. [PMID: 34502372 PMCID: PMC8430486 DOI: 10.3390/ijms22179464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/06/2021] [Indexed: 12/20/2022] Open
Abstract
Current research suggests that alterations in neurodevelopmental processes, involving gene X environment interactions during key stages of brain development (prenatal period and adolescence), are a major risk for schizophrenia. First, epidemiological studies supporting a genetic contribution to schizophrenia are presented in this article, including family, twin, and adoption studies. Then, an extensive literature review on genetic disorders associated with schizophrenia is reviewed. These epidemiological findings and clinical observations led researchers to conduct studies on genetic associations in schizophrenia, and more specifically on genomics (CNV: copy-number variant, and SNP: single nucleotide polymorphism). The main structural (CNV) and sequence (SNP) variants found in individuals with schizophrenia are reported here. Evidence of genetic contributions to schizophrenia and current knowledge on genetic syndromes associated with this psychiatric disorder highlight the importance of a clinical genetic examination to detect minor physical anomalies in individuals with ultra-high risk of schizophrenia. Several dysmorphic features have been described in schizophrenia, especially in early onset schizophrenia, and can be viewed as neurodevelopmental markers of vulnerability. Early detection of individuals with neurodevelopmental abnormalities is a fundamental issue to develop prevention and diagnostic strategies, therapeutic intervention and follow-up, and to ascertain better the underlying mechanisms involved in the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Anne-Clémence Priol
- Pôle Hospitalo-Universitaire de Psychiatrie de l’Enfant et de l’Adolescent (PHUPEA), Centre Hospitalier Guillaume Régnier, University of Rennes 1, 35000 Rennes, France; (L.D.); (G.B.); (M.T.)
- Correspondence: (A.-C.P.); (S.T.); Tel.: +33-2-99-51-06-04 (A.-C.P. & S.T.); Fax: +33-2-99-32-46-98 (A.-C.P. & S.T.)
| | - Laure Denis
- Pôle Hospitalo-Universitaire de Psychiatrie de l’Enfant et de l’Adolescent (PHUPEA), Centre Hospitalier Guillaume Régnier, University of Rennes 1, 35000 Rennes, France; (L.D.); (G.B.); (M.T.)
| | - Gaella Boulanger
- Pôle Hospitalo-Universitaire de Psychiatrie de l’Enfant et de l’Adolescent (PHUPEA), Centre Hospitalier Guillaume Régnier, University of Rennes 1, 35000 Rennes, France; (L.D.); (G.B.); (M.T.)
| | - Mathieu Thépaut
- Pôle Hospitalo-Universitaire de Psychiatrie de l’Enfant et de l’Adolescent (PHUPEA), Centre Hospitalier Guillaume Régnier, University of Rennes 1, 35000 Rennes, France; (L.D.); (G.B.); (M.T.)
| | - Marie-Maude Geoffray
- Department of Child and Adolescent Psychiatry, Centre Hospitalier Le Vinatier, 69500 Bron, France;
| | - Sylvie Tordjman
- Pôle Hospitalo-Universitaire de Psychiatrie de l’Enfant et de l’Adolescent (PHUPEA), Centre Hospitalier Guillaume Régnier, University of Rennes 1, 35000 Rennes, France; (L.D.); (G.B.); (M.T.)
- CIC (Clinical Investigation Center) 1414 Inserm, Centre Hospitalier Universitaire (CHU) de Rennes, University of Rennes 1, 35033 Rennes, France
- Integrative Neuroscience and Cognition Center (INCC), CNRS UMR 8002, University of Paris, 75006 Paris, France
- Correspondence: (A.-C.P.); (S.T.); Tel.: +33-2-99-51-06-04 (A.-C.P. & S.T.); Fax: +33-2-99-32-46-98 (A.-C.P. & S.T.)
| |
Collapse
|
16
|
Is NRXN1 Gene Expression an Important Marker of Treatment of Depressive Disorders? A Pilot Study. J Pers Med 2021; 11:jpm11070637. [PMID: 34357104 PMCID: PMC8303668 DOI: 10.3390/jpm11070637] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/07/2021] [Accepted: 07/01/2021] [Indexed: 11/17/2022] Open
Abstract
Aim: Due to the fact that NRXN1 is associated with neurodevelopmental disorders, the aim of this study was to investigate the role of the NRXN1 gene in the etiology and epigenetics of depression by comparison of NRXN1 mRNA expression and NRXN1 protein level expression in patients suffering from depression versus healthy controls, as well as to search for clinical variables related to expression of the analyzed gene. Material and Methods: A total of 180 people aged 19–64 qualified for the study. The experimental group consisted of 97 people who were psychiatrically hospitalized, diagnosed with recurrent depressive disorders (F33) or who met the diagnostic criteria of a depressive episode (F32) according to ICD-10. The control group included 83 healthy people who volunteered to participate in the study. A sample of peripheral blood was obtained from people who were positively qualified to participate in the study—twice in the experimental group and once in the control group for genetic testing. Sociodemographic variables and data on the course of the disorder were also gathered. Patients were examined on study entry and at the end of the hospitalization with the Hamilton Depression Scale. Obtained data were analyzed statistically. The study was approved by the University’s Bioethics Committee. Results: The gene expression of NRXN1 at both mRNA and protein level significantly differs and it is lower in the experimental group compared to expression in healthy people. The difference in gene expression of NRXN1 at both the mRNA and protein levels between the first and second measurement in the experimental group is also significant. The result demonstrates a higher expression level in the first measurement and lower expression level in the second measurement when reported depression symptoms are less severe. Conclusions: Results concerning expression of NRXN1 may play an important role in further researches about the etiopathogenesis of depressive disorders such as looking for depression biomarkers and identifying evidence which may be relevant to personalize treatment for depression.
Collapse
|
17
|
Davatolhagh MF, Fuccillo MV. Neurexin1⍺ differentially regulates synaptic efficacy within striatal circuits. Cell Rep 2021; 34:108773. [PMID: 33626349 PMCID: PMC8071350 DOI: 10.1016/j.celrep.2021.108773] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 11/18/2020] [Accepted: 01/29/2021] [Indexed: 02/08/2023] Open
Abstract
Mutations in genes essential for synaptic function, such as the presynaptic adhesion molecule Neurexin1α (Nrxn1α), are strongly implicated in neuropsychiatric pathophysiology. As the input nucleus of the basal ganglia, the striatum integrates diverse excitatory projections governing cognitive and motor control, and its impairment may represent a recurrent pathway to disease. Here, we test the functional relevance of Nrxn1α in striatal circuits by employing optogenetic-mediated afferent recruitment of dorsal prefrontal cortical (dPFC) and parafascicular thalamic connections onto dorsomedial striatal (DMS) spiny projection neurons (SPNs). For dPFC-DMS circuits, we find decreased synaptic strength specifically onto indirect pathway SPNs in both Nrxn1α+/- and Nrxn1α-/- mice, driven by reductions in neurotransmitter release. In contrast, thalamic excitatory inputs to DMS exhibit relatively normal excitatory synaptic strength despite changes in synaptic N-methyl-D-aspartate receptor (NMDAR) content. These findings suggest that dysregulation of Nrxn1α modulates striatal function in an input- and target-specific manner.
Collapse
Affiliation(s)
- M Felicia Davatolhagh
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marc V Fuccillo
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
18
|
Chowdhury D, Watters K, Biederer T. Synaptic recognition molecules in development and disease. Curr Top Dev Biol 2021; 142:319-370. [PMID: 33706921 DOI: 10.1016/bs.ctdb.2020.12.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Synaptic connectivity patterns underlie brain functions. How recognition molecules control where and when neurons form synapses with each other, therefore, is a fundamental question of cellular neuroscience. This chapter delineates adhesion and signaling complexes as well as secreted factors that contribute to synaptic partner recognition in the vertebrate brain. The sections follow a developmental perspective and discuss how recognition molecules (1) guide initial synaptic wiring, (2) provide for the rejection of incorrect partner choices, (3) contribute to synapse specification, and (4) support the removal of inappropriate synapses once formed. These processes involve a rich repertoire of molecular players and key protein families are described, notably the Cadherin and immunoglobulin superfamilies, Semaphorins/Plexins, Leucine-rich repeat containing proteins, and Neurexins and their binding partners. Molecular themes that diversify these recognition systems are defined and highlighted throughout the text, including the neuron-type specific expression and combinatorial action of recognition factors, alternative splicing, and post-translational modifications. Methodological innovations advancing the field such as proteomic approaches and single cell expression studies are additionally described. Further, the chapter highlights the importance of choosing an appropriate brain region to analyze synaptic recognition factors and the advantages offered by laminated structures like the hippocampus or retina. In a concluding section, the profound disease relevance of aberrant synaptic recognition for neurodevelopmental and psychiatric disorders is discussed. Based on the current progress, an outlook is presented on research goals that can further advance insights into how recognition molecules provide for the astounding precision and diversity of synaptic connections.
Collapse
Affiliation(s)
| | - Katherine Watters
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States; Neuroscience Graduate Program, Tufts University School of Medicine, Boston, MA, United States
| | - Thomas Biederer
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States.
| |
Collapse
|
19
|
Thygesen JH, Presman A, Harju-Seppänen J, Irizar H, Jones R, Kuchenbaecker K, Lin K, Alizadeh BZ, Austin-Zimmerman I, Bartels-Velthuis A, Bhat A, Bruggeman R, Cahn W, Calafato S, Crespo-Facorro B, de Haan L, de Zwarte SMC, Di Forti M, Díez-Revuelta Á, Hall J, Hall MH, Iyegbe C, Jablensky A, Kahn R, Kalaydjieva L, Kravariti E, Lawrie S, Luykx JJ, Mata I, McDonald C, McIntosh AM, McQuillin A, Muir R, Ophoff R, Picchioni M, Prata DP, Ranlund S, Rujescu D, Rutten BPF, Schulze K, Shaikh M, Schirmbeck F, Simons CJP, Toulopoulou T, van Amelsvoort T, van Haren N, van Os J, van Winkel R, Vassos E, Walshe M, Weisbrod M, Zartaloudi E, Bell V, Powell J, Lewis CM, Murray RM, Bramon E. Genetic copy number variants, cognition and psychosis: a meta-analysis and a family study. Mol Psychiatry 2021; 26:5307-5319. [PMID: 32719466 PMCID: PMC8589646 DOI: 10.1038/s41380-020-0820-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 06/11/2020] [Accepted: 06/11/2020] [Indexed: 02/06/2023]
Abstract
The burden of large and rare copy number genetic variants (CNVs) as well as certain specific CNVs increase the risk of developing schizophrenia. Several cognitive measures are purported schizophrenia endophenotypes and may represent an intermediate point between genetics and the illness. This paper investigates the influence of CNVs on cognition. We conducted a systematic review and meta-analysis of the literature exploring the effect of CNV burden on general intelligence. We included ten primary studies with a total of 18,847 participants and found no evidence of association. In a new psychosis family study, we investigated the effects of CNVs on specific cognitive abilities. We examined the burden of large and rare CNVs (>200 kb, <1% MAF) as well as known schizophrenia-associated CNVs in patients with psychotic disorders, their unaffected relatives and controls (N = 3428) from the Psychosis Endophenotypes International Consortium (PEIC). The carriers of specific schizophrenia-associated CNVs showed poorer performance than non-carriers in immediate (P = 0.0036) and delayed (P = 0.0115) verbal recall. We found suggestive evidence that carriers of schizophrenia-associated CNVs had poorer block design performance (P = 0.0307). We do not find any association between CNV burden and cognition. Our findings show that the known high-risk CNVs are not only associated with schizophrenia and other neurodevelopmental disorders, but are also a contributing factor to impairment in cognitive domains such as memory and perceptual reasoning, and act as intermediate biomarkers of disease risk.
Collapse
Affiliation(s)
- Johan H. Thygesen
- grid.83440.3b0000000121901201Division of Psychiatry, University College London, London, UK
| | - Amelia Presman
- grid.83440.3b0000000121901201Division of Psychiatry, University College London, London, UK
| | - Jasmine Harju-Seppänen
- grid.83440.3b0000000121901201Division of Psychiatry, University College London, London, UK
| | - Haritz Irizar
- grid.83440.3b0000000121901201Division of Psychiatry, University College London, London, UK
| | - Rebecca Jones
- grid.83440.3b0000000121901201Division of Psychiatry, University College London, London, UK
| | - Karoline Kuchenbaecker
- grid.83440.3b0000000121901201Division of Psychiatry, University College London, London, UK ,grid.83440.3b0000000121901201UCL Genetics Institute, University College London, London, UK
| | - Kuang Lin
- grid.13097.3c0000 0001 2322 6764Institute of Psychiatry, Psychology & Neuroscience at King’s College London, London, UK ,grid.4991.50000 0004 1936 8948Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Behrooz Z. Alizadeh
- grid.4494.d0000 0000 9558 4598University of Groningen, University Medical Center Groningen, University Center for Psychiatry, Rob Giel Research Center, Groningen, The Netherlands ,grid.4494.d0000 0000 9558 4598Department of Epidemiology, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Agna Bartels-Velthuis
- grid.4494.d0000 0000 9558 4598University of Groningen, University Medical Center Groningen, University Center for Psychiatry, Rob Giel Research Center, Groningen, The Netherlands
| | - Anjali Bhat
- grid.83440.3b0000000121901201Division of Psychiatry, University College London, London, UK
| | - Richard Bruggeman
- grid.4494.d0000 0000 9558 4598University of Groningen, University Medical Center Groningen, University Center for Psychiatry, Rob Giel Research Center, Groningen, The Netherlands ,grid.4830.f0000 0004 0407 1981Department of Clinical and Developmental Neuropsychology, University of Groningen, Groningen, The Netherlands
| | - Wiepke Cahn
- grid.5477.10000000120346234University Medical Center Utrecht, Department of Psychiatry, Brain Centre Rudolf Magnus, Utrecht University, Utrecht, The Netherlands ,grid.413664.2Altrecht, General Mental Health Care, Utrecht, The Netherlands
| | - Stella Calafato
- grid.83440.3b0000000121901201Division of Psychiatry, University College London, London, UK
| | - Benedicto Crespo-Facorro
- grid.469673.90000 0004 5901 7501CIBERSAM, Centro Investigación Biomédica en Red Salud Mental, Sevilla, Spain ,grid.7821.c0000 0004 1770 272XUniversity Hospital Marqués de Valdecilla, University of Cantabria–IDIVAL, Santander, Spain ,grid.9224.d0000 0001 2168 1229Hospital Universitario Virgen del Rocío, IBiS, Department of Psychiatry, School of Medicine, University of Sevilla, Sevilla, Spain
| | - Liewe de Haan
- grid.7177.60000000084992262Amsterdam UMC, Department of Psychiatry, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands ,grid.491093.60000 0004 0378 2028Arkin, Institute for Mental Health, Amsterdam, The Netherlands
| | - Sonja M. C. de Zwarte
- grid.5477.10000000120346234University Medical Center Utrecht, Department of Psychiatry, Brain Centre Rudolf Magnus, Utrecht University, Utrecht, The Netherlands
| | - Marta Di Forti
- grid.13097.3c0000 0001 2322 6764Institute of Psychiatry, Psychology & Neuroscience at King’s College London, London, UK
| | - Álvaro Díez-Revuelta
- grid.83440.3b0000000121901201Division of Psychiatry, University College London, London, UK ,grid.5690.a0000 0001 2151 2978Laboratory of Cognitive and Computational Neuroscience—Centre for Biomedical Technology (CTB), Complutense University and Technical University of Madrid, Madrid, Spain
| | - Jeremy Hall
- grid.5600.30000 0001 0807 5670School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, UK
| | - Mei-Hua Hall
- grid.38142.3c000000041936754XPsychosis Neurobiology Laboratory, Harvard Medical School, McLean Hospital, Belmont, MA USA
| | - Conrad Iyegbe
- grid.13097.3c0000 0001 2322 6764Institute of Psychiatry, Psychology & Neuroscience at King’s College London, London, UK
| | - Assen Jablensky
- grid.1012.20000 0004 1936 7910Centre for Clinical Research in Neuropsychiatry, The University of Western Australia, Perth, WA Australia
| | - Rene Kahn
- grid.5477.10000000120346234University Medical Center Utrecht, Department of Psychiatry, Brain Centre Rudolf Magnus, Utrecht University, Utrecht, The Netherlands ,grid.59734.3c0000 0001 0670 2351Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Luba Kalaydjieva
- grid.1012.20000 0004 1936 7910Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Perth, WA Australia
| | - Eugenia Kravariti
- grid.13097.3c0000 0001 2322 6764Institute of Psychiatry, Psychology & Neuroscience at King’s College London, London, UK
| | - Stephen Lawrie
- grid.4305.20000 0004 1936 7988Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, Scotland UK
| | - Jurjen J. Luykx
- grid.5477.10000000120346234University Medical Center Utrecht, Department of Psychiatry, Brain Centre Rudolf Magnus, Utrecht University, Utrecht, The Netherlands ,grid.7692.a0000000090126352Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.491146.f0000 0004 0478 3153Second opinion outpatient clinic, GGNet Mental Health, Warsnveld, The Netherlands
| | - Igancio Mata
- grid.469673.90000 0004 5901 7501CIBERSAM, Centro Investigación Biomédica en Red Salud Mental, Sevilla, Spain ,Fundación Argibide, Pamplona, Spain
| | - Colm McDonald
- grid.6142.10000 0004 0488 0789The Centre for Neuroimaging & Cognitive Genomics (NICOG) and NCBES Galway Neuroscience Centre, National University of Ireland Galway, Galway, Ireland
| | - Andrew M. McIntosh
- grid.4305.20000 0004 1936 7988Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, Scotland UK ,grid.4305.20000 0004 1936 7988Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Andrew McQuillin
- grid.83440.3b0000000121901201Division of Psychiatry, University College London, London, UK
| | - Rebecca Muir
- grid.83440.3b0000000121901201Division of Psychiatry, University College London, London, UK
| | - Roel Ophoff
- grid.19006.3e0000 0000 9632 6718Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA USA ,grid.5645.2000000040459992XDepartment of Psychiatry, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marco Picchioni
- grid.13097.3c0000 0001 2322 6764Institute of Psychiatry, Psychology & Neuroscience at King’s College London, London, UK
| | - Diana P. Prata
- grid.13097.3c0000 0001 2322 6764Institute of Psychiatry, Psychology & Neuroscience at King’s College London, London, UK ,grid.9983.b0000 0001 2181 4263Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciencias da Universidade de Lisboa, Lisboa, Portugal ,grid.45349.3f0000 0001 2220 8863Centre for Psychological Research and Social Intervention, ISCTE-IUL, Lisboa, Portugal
| | - Siri Ranlund
- grid.83440.3b0000000121901201Division of Psychiatry, University College London, London, UK ,grid.13097.3c0000 0001 2322 6764Institute of Psychiatry, Psychology & Neuroscience at King’s College London, London, UK
| | - Dan Rujescu
- grid.5252.00000 0004 1936 973XDepartment of Psychiatry, Ludwig-Maximilians University of Munich, Munich, Germany ,grid.9018.00000 0001 0679 2801Department of Psychiatry, Psychotherapy and Psychosomatics, University of Halle Wittenberg, Halle, Germany
| | - Bart P. F. Rutten
- grid.412966.e0000 0004 0480 1382Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, The Netherlands ,grid.412966.e0000 0004 0480 1382The Brain+Nerve Centre, Maastricht University Medical Centre+ (MUMC+), Maastricht, The Netherlands
| | - Katja Schulze
- grid.13097.3c0000 0001 2322 6764Institute of Psychiatry, Psychology & Neuroscience at King’s College London, London, UK ,grid.37640.360000 0000 9439 0839South London and Maudsley NHS Foundation Trust, London, UK
| | - Madiha Shaikh
- grid.451079.e0000 0004 0428 0265North East London Foundation Trust, London, UK ,grid.83440.3b0000000121901201Research Department of Clinical, Educational and Health Psychology, University College London, London, UK
| | - Frederike Schirmbeck
- grid.7177.60000000084992262Amsterdam UMC, Department of Psychiatry, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands ,grid.491093.60000 0004 0378 2028Arkin, Institute for Mental Health, Amsterdam, The Netherlands
| | - Claudia J. P. Simons
- grid.412966.e0000 0004 0480 1382Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, The Netherlands ,grid.491104.9GGzE Institute for Mental Health Care, Eindhoven, The Netherlands
| | - Timothea Toulopoulou
- grid.13097.3c0000 0001 2322 6764Institute of Psychiatry, Psychology & Neuroscience at King’s College London, London, UK ,grid.18376.3b0000 0001 0723 2427Department of Psychology, Bilkent University, Main Campus, Bilkent, Ankara Turkey
| | - Therese van Amelsvoort
- grid.412966.e0000 0004 0480 1382Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Neeltje van Haren
- grid.5477.10000000120346234University Medical Center Utrecht, Department of Psychiatry, Brain Centre Rudolf Magnus, Utrecht University, Utrecht, The Netherlands ,grid.5645.2000000040459992XDepartment of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, Sophia’s Children Hospital, Rotterdam, The Netherlands
| | - Jim van Os
- grid.13097.3c0000 0001 2322 6764Institute of Psychiatry, Psychology & Neuroscience at King’s College London, London, UK ,grid.412966.e0000 0004 0480 1382Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, The Netherlands ,grid.7692.a0000000090126352Department of Psychiatry, UMC Utrecht Brain Center, Utrecht, The Netherlands
| | - Ruud van Winkel
- grid.5596.f0000 0001 0668 7884KU Leuven, Department of Neuroscience, Research Group Psychiatry, Leuven, Belgium
| | - Evangelos Vassos
- grid.13097.3c0000 0001 2322 6764Institute of Psychiatry, Psychology & Neuroscience at King’s College London, London, UK
| | - Muriel Walshe
- grid.83440.3b0000000121901201Division of Psychiatry, University College London, London, UK
| | - Matthias Weisbrod
- grid.7700.00000 0001 2190 4373Department of General Psychiatry, Center of Psychosocial Medicine, University of Heidelberg, Heidelberg, Germany ,grid.490718.30000000406368535SRH Klinikum, Karlsbad-Langensteinbach, Germany
| | - Eirini Zartaloudi
- grid.83440.3b0000000121901201Division of Psychiatry, University College London, London, UK
| | - Vaughan Bell
- grid.83440.3b0000000121901201Division of Psychiatry, University College London, London, UK
| | - John Powell
- grid.13097.3c0000 0001 2322 6764Institute of Psychiatry, Psychology & Neuroscience at King’s College London, London, UK
| | - Cathryn M. Lewis
- grid.13097.3c0000 0001 2322 6764Institute of Psychiatry, Psychology & Neuroscience at King’s College London, London, UK
| | - Robin M. Murray
- grid.13097.3c0000 0001 2322 6764Institute of Psychiatry, Psychology & Neuroscience at King’s College London, London, UK ,grid.37640.360000 0000 9439 0839South London and Maudsley NHS Foundation Trust, London, UK
| | - Elvira Bramon
- Division of Psychiatry, University College London, London, UK. .,Institute of Psychiatry, Psychology & Neuroscience at King's College London, London, UK. .,Institute of Cognitive Neuroscience, University College London, London, UK.
| |
Collapse
|
20
|
Neurexin 1 variants as risk factors for suicide death. Mol Psychiatry 2021; 26:7436-7445. [PMID: 34168285 PMCID: PMC8709873 DOI: 10.1038/s41380-021-01190-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/20/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023]
Abstract
Suicide is a significant public health concern with complex etiology. Although the genetic component of suicide is well established, the scope of gene networks and biological mechanisms underlying suicide has yet to be defined. Previously, we reported genome-wide evidence that neurexin 1 (NRXN1), a key synapse organizing molecule, is associated with familial suicide risk. Here we present new evidence for two non-synonymous variants (rs78540316; P469S and rs199784139; H885Y) associated with increased familial risk of suicide death. We tested the impact of these variants on binding interactions with known partners and assessed functionality in a hemi-synapse formation assay. Although the formation of hemi-synapses was not altered with the P469S variant relative to wild-type, both variants increased binding to the postsynaptic binding partner, leucine-rich repeat transmembrane neuronal 2 (LRRTM2) in vitro. Our findings indicate that variants in NRXN1 and related synaptic genes warrant further study as risk factors for suicide death.
Collapse
|
21
|
Lee PH, Feng YCA, Smoller JW. Pleiotropy and Cross-Disorder Genetics Among Psychiatric Disorders. Biol Psychiatry 2021; 89:20-31. [PMID: 33131714 PMCID: PMC7898275 DOI: 10.1016/j.biopsych.2020.09.026] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/28/2020] [Accepted: 09/30/2020] [Indexed: 12/20/2022]
Abstract
Genome-wide analyses of common and rare genetic variations have documented the heritability of major psychiatric disorders, established their highly polygenic genetic architecture, and identified hundreds of contributing variants. In recent years, these studies have illuminated another key feature of the genetic basis of psychiatric disorders: the important role and pervasive nature of pleiotropy. It is now clear that a substantial fraction of genetic influences on psychopathology transcend clinical diagnostic boundaries. In this review, we summarize evidence in psychiatry for pleiotropy at multiple levels of analysis: from overall genome-wide correlation to biological pathways and down to the level of individual loci. We examine underlying mechanisms of observed pleiotropy, including genetic effects on neurodevelopment, diverse actions of regulatory elements, mediated effects, and spurious associations of genomic variation with multiple phenotypes. We conclude with an exploration of the implications of pleiotropy for understanding the genetic basis of psychiatric disorders, informing nosology, and advancing the aims of precision psychiatry and genomic medicine.
Collapse
Affiliation(s)
- Phil H Lee
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, and Department of Psychiatry, Massachusetts General Hospital, Boston; and Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Yen-Chen A Feng
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, and Department of Psychiatry, Massachusetts General Hospital, Boston; and Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Jordan W Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, and Department of Psychiatry, Massachusetts General Hospital, Boston; and Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts.
| |
Collapse
|
22
|
Alabi OO, Davatolhagh MF, Robinson M, Fortunato MP, Vargas Cifuentes L, Kable JW, Fuccillo MV. Disruption of Nrxn1α within excitatory forebrain circuits drives value-based dysfunction. eLife 2020; 9:e54838. [PMID: 33274715 PMCID: PMC7759380 DOI: 10.7554/elife.54838] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 12/03/2020] [Indexed: 01/17/2023] Open
Abstract
Goal-directed behaviors are essential for normal function and significantly impaired in neuropsychiatric disorders. Despite extensive associations between genetic mutations and these disorders, the molecular contributions to goal-directed dysfunction remain unclear. We examined mice with constitutive and brain region-specific mutations in Neurexin1α, a neuropsychiatric disease-associated synaptic molecule, in value-based choice paradigms. We found Neurexin1α knockouts exhibited reduced selection of beneficial outcomes and impaired avoidance of costlier options. Reinforcement modeling suggested that this was driven by deficits in updating and representation of value. Disruption of Neurexin1α within telencephalic excitatory projection neurons, but not thalamic neurons, recapitulated choice abnormalities of global Neurexin1α knockouts. Furthermore, this selective forebrain excitatory knockout of Neurexin1α perturbed value-modulated neural signals within striatum, a central node in feedback-based reinforcement learning. By relating deficits in value-based decision-making to region-specific Nrxn1α disruption and changes in value-modulated neural activity, we reveal potential neural substrates for the pathophysiology of neuropsychiatric disease-associated cognitive dysfunction.
Collapse
Affiliation(s)
- Opeyemi O Alabi
- Department of NeurosciencePhiladelphiaUnited States
- Neuroscience Graduate Group, Perelman School of MedicinePhiladelphiaUnited States
| | - M Felicia Davatolhagh
- Department of NeurosciencePhiladelphiaUnited States
- Neuroscience Graduate Group, Perelman School of MedicinePhiladelphiaUnited States
| | | | | | - Luigim Vargas Cifuentes
- Department of NeurosciencePhiladelphiaUnited States
- Neuroscience Graduate Group, Perelman School of MedicinePhiladelphiaUnited States
| | - Joseph W Kable
- Department of Psychology, University of PennsylvaniaPhiladelphiaUnited States
| | | |
Collapse
|
23
|
Deletion of NRXN1α impairs long-range and local connectivity in amygdala fear circuit. Transl Psychiatry 2020; 10:242. [PMID: 32684634 PMCID: PMC7370229 DOI: 10.1038/s41398-020-00926-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/07/2020] [Accepted: 07/09/2020] [Indexed: 01/26/2023] Open
Abstract
Neurexins are a family of presynaptic cell adhesion proteins that regulate synaptic structure and maintain normal synaptic transmission. Mutations in the α-isoform of neurexin1-gene (NRXN1α) are linked with cognitive and emotional dysregulation, which are heavily dependent on the amygdala and medial prefrontal cortex (mPFC). It is however not known whether deletion of NRXN1α gene affect specific synaptic elements within the amygdala microcircuit and connectivity with mPFC. In this study, we show that NRXN1α deletion impairs synaptic transmission between the dorsal medial prefrontal cortex (dmPFC) and basal amygdala (BA) principal neurons. Stimulation of dmPFC fibers resulted in reduced paired pulse ratio (PPR) and AMPA/NMDA ratio at dmPFC to BA synapses in NRXN1α-knockout (KO) (NRXN1α KO) mice suggestive of pre- and postsynaptic deficits but there was no change at the lateral amygdala (LA) to BA synapses following LA stimulation. However, feedforward inhibition from either pathway was significantly reduced, suggestive of input-independent deficit in GABAergic transmission within BA. We further analyzed BA inhibitory network and found reduced connectivity between BA GABAergic and glutamatergic neurons in NRXN1α KO mice. As this circuit is tightly linked with fear regulation, we subjected NRXN1α KO and WT mice to discriminative fear conditioning and found a deficit in fear memory retrieval in NRXN1α KO mice compared with WT mice. Together, we provide novel evidence that deletion of NRNX1α disrupts amygdala fear circuit.
Collapse
|
24
|
Furlong MA, Paul KC, Yan Q, Chuang YH, Cockburn MG, Bronstein JM, Horvath S, Ritz B. An epigenome-wide association study of ambient pyrethroid pesticide exposures in California's central valley. Int J Hyg Environ Health 2020; 229:113569. [PMID: 32679516 DOI: 10.1016/j.ijheh.2020.113569] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 05/08/2020] [Accepted: 05/22/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Pyrethroid pesticide use is increasing worldwide, although the full extent of associated health effects is unknown. An epigenome-wide association study (EWAS) with exploratory pathway analysis may help identify potential pyrethroid-related health effects. METHODS We performed an exploratory EWAS of chronic ambient pyrethroid exposure using control participants' blood in the Parkinson's Environment and Genes Study in the Central Valley of California (N = 237). We estimated associations of living and working near agricultural pyrethroid pesticide applications in the past 5 years (binary) with site-specific differential methylation, and used a false discovery rate (FDR) cut off of 0.05 for significance. We controlled for age, sex, education, cell count, and an ancestral marker for Hispanic ethnicity. We normalized methylation values for Type I/II probe bias using Beta-Mixture Quantile (BMIQ) normalization, filtered out cross-reactive probes, and evaluated for remaining bias with Surrogate Variable Analysis (SVA). We also evaluated the effects of controlling for cell count and normalizing for Type I/II probe bias by comparing changes in effect estimates and p-values for the top hits across BMIQ and GenomeStudio normalization methods, and controlling for cell count. To facilitate broader interpretation, we annotated genes to the CpG sites and performed gene set overrepresentation analysis, using genes annotated to CpG sites that were associated with pyrethroids at a raw p < 0.05, and controlling for background representation of CpG sites on the chip. We did this for both a biological process context (Gene Ontology terms) using missMethyl, and a disease set context using WebGestalt. For these gene set overrepresentation analyses we also used an FDR cut off of 0.05 for significance of gene sets. RESULTS After controlling for cell count and applying BMIQ normalization, 4 CpG sites were differentially methylated in relation to pyrethroid exposures. When using GenomeStudio's Illumina normalization, 415 CpG sites were differentially methylated, including all four identified with the BMIQ method. In the gene set overrepresentation analyses, we identified 6 GO terms using BMIQ normalization, and 76 using Illumina normalization, including the 6 identified by BMIQ. For disease sets, we identified signals for Alzheimer's disease, leukemia and several other cancers, diabetes, birth defects, and other diseases, for both normalization methods. We identified minimal changes in effect estimates after controlling for cell count, and controlling for cell count generally weakened p-values. BMIQ normalization, however, resulted in different beta coefficients and weakened p-values. CONCLUSIONS Chronic ambient pyrethroid exposure is associated with differential methylation at CpG sites that annotate to a wide variety of disease states and biological mechanisms that align with prior research. However, this EWAS also implicates several novel diseases for future investigation, and highlights the relative importance of different background normalization methods in identifying associations.
Collapse
Affiliation(s)
- Melissa A Furlong
- Department of Community, Environment, and Policy, University of Arizona Mel and Enid Zuckerman College of Public Health, Tucson, AZ, USA.
| | - Kimberly C Paul
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Qi Yan
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Yu-Hsuan Chuang
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Myles G Cockburn
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, CA, USA
| | - Jeff M Bronstein
- Department of Neurology, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, Los Angeles, CA, USA; Department of Biostatistics, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Beate Ritz
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA; Department of Neurology, David Geffen School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
25
|
Li K, Li Y, Wang J, Huo Y, Huang D, Li S, Liu J, Li X, Liu R, Chen X, Yao YG, Chen C, Xiao X, Li M, Luo XJ. A functional missense variant in ITIH3 affects protein expression and neurodevelopment and confers schizophrenia risk in the Han Chinese population. J Genet Genomics 2020; 47:233-248. [PMID: 32712163 DOI: 10.1016/j.jgg.2020.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/30/2020] [Accepted: 04/02/2020] [Indexed: 12/12/2022]
Abstract
The Psychiatric Genomics Consortium (PGC) has recently identified 10 potential functional coding variants for schizophrenia. However, how these coding variants confer schizophrenia risk remains largely unknown. Here, we investigate the associations between eight potential functional coding variants identified by PGC and schizophrenia in a large Han Chinese sample (n = 4022 cases and 9270 controls). Among the eight tested single nucelotide polymorphisms (SNPs), rs3617 (a missense variant, p.K315Q in the ITIH3 gene) showed genome-wide significant association with schizophrenia in the Han Chinese population (P = 8.36 × 10-16), with the same risk allele as in PGC. Interestingly, rs3617 is located in a genomic region that is highly evolutionarily conserved, and its schizophrenia risk allele (C allele) was associated with lower ITIH3 mRNA and protein expression. Intriguingly, mouse neural stem cells stably overexpressing ITIH3 with different alleles of rs3617 exhibited significant differences in proliferation, migration, and differentiation, suggesting the impact of rs3617 on neurodevelopment. Subsequent transcriptome analysis found that the differentially expressed genes in neural stem cells stably overexpressing different alleles of rs3617 were significantly enriched in schizophrenia-related pathways, including cell adhesion, synapse assembly, MAPK and PI3K-AKT pathways. Our study provides convergent lines of evidence suggesting that rs3617 in ITIH3 likely affects protein function and neurodevelopment and thereby confers risk of schizophrenia.
Collapse
Affiliation(s)
- Kaiqin Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China
| | - Yifan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China
| | - Junyang Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China
| | - Yongxia Huo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Di Huang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Shiwu Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China
| | - Jiewei Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Xiaoyan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China
| | - Rong Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
| | - Xiaogang Chen
- Institute of Mental Health, National Clinical Research Center for Mental Health Disorders and National Technology Institute of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China; KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China; KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Xiong-Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China; KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.
| |
Collapse
|
26
|
Rees E, Owen MJ. Translating insights from neuropsychiatric genetics and genomics for precision psychiatry. Genome Med 2020; 12:43. [PMID: 32349784 PMCID: PMC7189552 DOI: 10.1186/s13073-020-00734-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 04/03/2020] [Indexed: 12/30/2022] Open
Abstract
The primary aim of precision medicine is to tailor healthcare more closely to the needs of individual patients. This requires progress in two areas: the development of more precise treatments and the ability to identify patients or groups of patients in the clinic for whom such treatments are likely to be the most effective. There is widespread optimism that advances in genomics will facilitate both of these endeavors. It can be argued that of all medical specialties psychiatry has most to gain in these respects, given its current reliance on syndromic diagnoses, the minimal foundation of existing mechanistic knowledge, and the substantial heritability of psychiatric phenotypes. Here, we review recent advances in psychiatric genomics and assess the likely impact of these findings on attempts to develop precision psychiatry. Emerging findings indicate a high degree of polygenicity and that genetic risk maps poorly onto the diagnostic categories used in the clinic. The highly polygenic and pleiotropic nature of psychiatric genetics will impact attempts to use genomic data for prediction and risk stratification, and also poses substantial challenges for conventional approaches to gaining biological insights from genetic findings. While there are many challenges to overcome, genomics is building an empirical platform upon which psychiatry can now progress towards better understanding of disease mechanisms, better treatments, and better ways of targeting treatments to the patients most likely to benefit, thus paving the way for precision psychiatry.
Collapse
Affiliation(s)
- Elliott Rees
- MRC Centre for Neuropsychiatric Genetics and Genomics, Neuroscience and Mental Health Research Institute and Division of Psychological Medicine and Clinical Neuroscience, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ UK
| | - Michael J. Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Neuroscience and Mental Health Research Institute and Division of Psychological Medicine and Clinical Neuroscience, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ UK
| |
Collapse
|
27
|
Szatkiewicz JP, Fromer M, Nonneman RJ, Ancalade N, Johnson JS, Stahl EA, Rees E, Bergen SE, Hultman CM, Kirov G, O'Donovan M, Owen M, Holmans P, Sklar P, Sullivan PF, Purcell SM, Crowley JJ, Ruderfer DM. Characterization of Single Gene Copy Number Variants in Schizophrenia. Biol Psychiatry 2020; 87:736-744. [PMID: 31767120 PMCID: PMC7103483 DOI: 10.1016/j.biopsych.2019.09.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 09/24/2019] [Accepted: 09/25/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND Genetic studies of schizophrenia have implicated numerous risk loci including several copy number variants (CNVs) of large effect and hundreds of loci of small effect. In only a few cases has a specific gene been clearly identified. Rare CNVs affecting a single gene offer a potential avenue to discovering schizophrenia risk genes. METHODS CNVs were generated from exome sequencing of 4913 schizophrenia cases and 6188 control subjects from Sweden. We integrated two CNV calling methods (XHMM and ExomeDepth) to expand our set of single-gene CNVs and leveraged two different approaches for validating these variants (quantitative polymerase chain reaction and NanoString). RESULTS We found a significant excess of all rare CNVs (deletions: p = .0004, duplications: p = .0006) and single-gene CNVs (deletions: p = .04, duplications: p = .03) in schizophrenia cases compared with control subjects. An expanded set of CNVs generated from integrating multiple approaches showed a significant burden of deletions in 11 of 21 gene sets previously implicated in schizophrenia and across all genes in those sets (p = .008), although no tests survived correction. We performed an extensive validation of all deletions in the significant set of voltage-gated calcium channels among CNVs called from both exome sequencing and genotyping arrays. In total, 4 exonic, single-gene deletions were validated in schizophrenia cases and none in control subjects (p = .039), of which all were identified by exome sequencing. CONCLUSIONS These results point to the potential contribution of single-gene CNVs to schizophrenia, indicate that the utility of exome sequencing for CNV calling has yet to be maximized, and note that single-gene CNVs should be included in gene-focused studies using other classes of variation.
Collapse
Affiliation(s)
- Jin P Szatkiewicz
- Center for Psychiatric Genomics, Department of Genetics and Psychiatry, University of North Carolina, Chapel Hill, North Carolina
| | - Menachem Fromer
- Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Randal J Nonneman
- Center for Psychiatric Genomics, Department of Genetics and Psychiatry, University of North Carolina, Chapel Hill, North Carolina
| | - NaEshia Ancalade
- Center for Psychiatric Genomics, Department of Genetics and Psychiatry, University of North Carolina, Chapel Hill, North Carolina
| | - Jessica S Johnson
- Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Eli A Stahl
- Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Elliott Rees
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Sarah E Bergen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Christina M Hultman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - George Kirov
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Michael O'Donovan
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Michael Owen
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Peter Holmans
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Pamela Sklar
- Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Patrick F Sullivan
- Center for Psychiatric Genomics, Department of Genetics and Psychiatry, University of North Carolina, Chapel Hill, North Carolina; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Shaun M Purcell
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - James J Crowley
- Center for Psychiatric Genomics, Department of Genetics and Psychiatry, University of North Carolina, Chapel Hill, North Carolina; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Douglas M Ruderfer
- Division of Genetic Medicine, Departments of Medicine, Psychiatry, and Biomedical Informatics, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
28
|
Jagannath V, Grünblatt E, Theodoridou A, Oneda B, Roth A, Gerstenberg M, Franscini M, Traber-Walker N, Correll CU, Heekeren K, Rössler W, Rauch A, Walitza S. Rare copy number variants in individuals at clinical high risk for psychosis: Enrichment of synaptic/brain-related functional pathways. Am J Med Genet B Neuropsychiatr Genet 2020; 183:140-151. [PMID: 31742845 DOI: 10.1002/ajmg.b.32770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 10/08/2019] [Accepted: 10/23/2019] [Indexed: 11/07/2022]
Abstract
Schizophrenia is a complex and chronic neuropsychiatric disorder, with a heritability of around 60-80%. Large (>100 kb) rare (<1%) copy number variants (CNVs) occur more frequently in schizophrenia patients compared to controls. Currently, there are no studies reporting genome-wide CNVs in clinical high risk for psychosis (CHR-P) individuals. The aim of this study was to investigate the role of rare genome-wide CNVs in 84 CHR-P individuals and 124 presumably healthy controls. There were no significant differences in all rare CNV frequencies and sizes between CHR-P individuals and controls. However, brain-related CNVs and brain-related deletions were significantly more frequent in CHR-P individuals than controls. In CHR-P individuals, significant associations were found between brain-related CNV carriers and attenuated positive symptoms syndrome or cognitive disturbances (OR = 3.07, p = .0286). Brain-related CNV carriers experienced significantly higher negative symptoms (p = .0047), higher depressive symptoms (p = .0175), and higher disturbances of self and surroundings (p = .0029) than noncarriers. Furthermore, enrichment analysis of genes was performed in the regions of rare CNVs using three independent methods, which confirmed significant clustering of predefined genes involved in synaptic/brain-related functional pathways in CHR-P individuals. These results suggest that rare CNVs might affect synaptic/brain-related functional pathways in CHR-P individuals.
Collapse
Affiliation(s)
- Vinita Jagannath
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry, University of Zurich, Zurich, Switzerland
| | - Edna Grünblatt
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Anastasia Theodoridou
- The Zurich Program for Sustainable Development of Mental Health Services (ZInEP), University Hospital of Psychiatry Zurich, Zurich, Switzerland.,Department of Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry, University of Zurich, Zurich, Switzerland
| | - Beatrice Oneda
- Institute of Medical Genetics, University of Zurich, Schlieren, Switzerland
| | - Alexander Roth
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry, University of Zurich, Zurich, Switzerland
| | - Miriam Gerstenberg
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry, University of Zurich, Zurich, Switzerland
| | - Maurizia Franscini
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry, University of Zurich, Zurich, Switzerland
| | - Nina Traber-Walker
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry, University of Zurich, Zurich, Switzerland
| | - Christoph U Correll
- Department of Psychiatry, The Zucker Hillside Hospital, Northwell Health, Glen Oaks, New York.,Department of Psychiatry and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York.,The Feinstein Institute for Medical Research, Manhasset, New York.,Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany
| | - Karsten Heekeren
- The Zurich Program for Sustainable Development of Mental Health Services (ZInEP), University Hospital of Psychiatry Zurich, Zurich, Switzerland.,Department of Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry, University of Zurich, Zurich, Switzerland
| | - Wulf Rössler
- The Zurich Program for Sustainable Development of Mental Health Services (ZInEP), University Hospital of Psychiatry Zurich, Zurich, Switzerland.,Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Anita Rauch
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.,Institute of Medical Genetics, University of Zurich, Schlieren, Switzerland
| | - Susanne Walitza
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.,The Zurich Program for Sustainable Development of Mental Health Services (ZInEP), University Hospital of Psychiatry Zurich, Zurich, Switzerland
| |
Collapse
|
29
|
Hart MP. Stress-Induced Neuron Remodeling Reveals Differential Interplay Between Neurexin and Environmental Factors in Caenorhabditis elegans. Genetics 2019; 213:1415-1430. [PMID: 31558583 PMCID: PMC6893388 DOI: 10.1534/genetics.119.302415] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 09/24/2019] [Indexed: 01/30/2023] Open
Abstract
Neurexins are neuronal adhesion molecules important for synapse maturation, function, and plasticity. Neurexins have been genetically associated with neurodevelopmental disorders, including autism spectrum disorders (ASDs) and schizophrenia, but can have variable penetrance and phenotypic severity. Heritability studies indicate that a significant percentage of risk for ASD and schizophrenia includes environmental factors, highlighting a poorly understood interplay between genetic and environmental factors. The singular Caenorhabditis elegans ortholog of human neurexins, nrx-1, controls experience-dependent morphologic remodeling of a GABAergic neuron in adult males. Here, I show remodeling of this neuron's morphology in response to each of three environmental stressors (nutritional, heat, or genotoxic stress) when applied specifically during sexual maturation. Increased outgrowth of axon-like neurites following adolescent stress is the result of an altered morphologic plasticity in adulthood. Despite remodeling being induced by each of the three stressors, only nutritional stress affects downstream behavior and is dependent on neurexin/nrx-1 Heat or genotoxic stress in adolescence does not alter behavior despite inducing GABAergic neuron remodeling, in a neurexin/nrx-1 independent fashion. Starvation-induced remodeling is also dependent on neuroligin/nlg-1, the canonical binding partner for neurexin/nrx-1, and the transcription factors FOXO/daf-16 and HSF1/hsf-1hsf-1 and daf-16, in addition, each have unique roles in remodeling induced by heat and UV stress. The differential molecular mechanisms underlying GABAergic neuron remodeling in response to different stressors, and the disparate effects of stressors on downstream behavior, are a paradigm for understanding how genetics, environmental exposures, and plasticity may contribute to brain dysfunction in ASDs and schizophrenia.
Collapse
Affiliation(s)
- Michael P Hart
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
30
|
Hu Z, Xiao X, Zhang Z, Li M. Genetic insights and neurobiological implications from NRXN1 in neuropsychiatric disorders. Mol Psychiatry 2019; 24:1400-1414. [PMID: 31138894 DOI: 10.1038/s41380-019-0438-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 03/31/2019] [Accepted: 04/29/2019] [Indexed: 02/08/2023]
Abstract
Many neuropsychiatric and neurodevelopmental disorders commonly share genetic risk factors. To date, the mechanisms driving the pathogenesis of these disorders, particularly how genetic variations affect the function of risk genes and contribute to disease symptoms, remain largely unknown. Neurexins are a family of synaptic adhesion molecules, which play important roles in the formation and establishment of synaptic structure, as well as maintenance of synaptic function. Accumulating genomic findings reveal that genetic variations within genes encoding neurexins are associated with a variety of psychiatric conditions such as schizophrenia, autism spectrum disorder, and some developmental abnormalities. In this review, we focus on NRXN1, one of the most compelling psychiatric risk genes of the neurexin family. We performed a comprehensive survey and analysis of current genetic and molecular data including both common and rare alleles within NRXN1 associated with psychiatric illnesses, thus providing insights into the genetic risk conferred by NRXN1. We also summarized the neurobiological evidences, supporting the function of NRXN1 and its protein products in synaptic formation, organization, transmission and plasticity, as well as disease-relevant behaviors, and assessed the mechanistic link between the mutations of NRXN1 and synaptic and behavioral pathology in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Zhonghua Hu
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China. .,Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China. .,Department of Psychiatry, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China. .,National Clinical Research Center on Mental Disorders, Changsha, Hunan, China.
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Zhuohua Zhang
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China. .,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
31
|
FunVar: A systematic pipeline to unravel the convergence patterns of genetic variants in ASD, a paradigmatic complex disease. J Biomed Inform 2019; 98:103273. [PMID: 31454647 DOI: 10.1016/j.jbi.2019.103273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 07/28/2019] [Accepted: 08/24/2019] [Indexed: 11/22/2022]
Abstract
In recent years, the technological advances for capturing genetic variation in large populations led to the identification of large numbers of putative or disease-causing variants. However, their mechanistic understanding is lagging far behind and has posed new challenges regarding their relevance for disease phenotypes, particularly for common complex disorders. In this study, we propose a systematic pipeline to infer biological meaning from genetic variants, namely rare Copy Number Variants (CNVs). The pipeline consists of three modules that seek to (1) improve genetic data quality by excluding low confidence CNVs, (2) identify disrupted biological processes, and (3) aggregate similar enriched biological processes terms using semantic similarity. The proposed pipeline was applied to CNVs from individuals diagnosed with Autism Spectrum Disorder (ASD). We found that rare CNVs disrupting brain expressed genes dysregulated a wide range of biological processes, such as nervous system development and protein polyubiquitination. The disrupted biological processes identified in ASD patients were in accordance with previous findings. This coherence with literature indicates the feasibility of the proposed pipeline in interpreting the biological role of genetic variants in complex disease development. The suggested pipeline is easily adjustable at each step and its independence from any specific dataset and software makes it an effective tool in analyzing existing genetic resources. The FunVar pipeline is available at https://github.com/lasigeBioTM/FunVar and includes pre and post processing steps to effectively interpret biological mechanisms of putative disease causing genetic variants.
Collapse
|
32
|
Moslem M, Olive J, Falk A. Stem cell models of schizophrenia, what have we learned and what is the potential? Schizophr Res 2019; 210:3-12. [PMID: 30587427 DOI: 10.1016/j.schres.2018.12.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 12/14/2018] [Accepted: 12/16/2018] [Indexed: 12/13/2022]
Abstract
Schizophrenia is a complex disorder with clinical manifestations in early adulthood. However, it may start with disruption of brain development caused by genetic or environmental factors, or both. Early deteriorating effects of genetic/environmental factors on neural development might be key to described disease causing mechanisms. Establishing cellular models with cells from affected individual using the induced pluripotent stem cells (iPSC) technology could be used to mimic early neurodevelopment alterations caused by risk genes or environmental stressors. Indeed, cellular models have allowed identification and further study of risk factors and the biological pathways in which they are involved. New advancements in differentiation methods such as defined and robust monolayer protocols and cerebral 3D organoids have made it possible to faithfully mimic neural development and neuronal functionality while CRISPR-editing tools assist to engineer isogenic cell lines to precisely explore genetic variation in polygenic diseases such as schizophrenia. Here we review the current field of iPSC models of schizophrenia and how risk factors can be modelled as well as discussing the common biological pathways involved.
Collapse
Affiliation(s)
- Mohsen Moslem
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Jessica Olive
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Life Sciences, Imperial College London, United Kingdom.
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
33
|
Sriretnakumar V, Zai CC, Wasim S, Barsanti-Innes B, Kennedy JL, So J. Copy number variant syndromes are frequent in schizophrenia: Progressing towards a CNV-schizophrenia model. Schizophr Res 2019; 209:171-178. [PMID: 31080157 DOI: 10.1016/j.schres.2019.04.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 02/26/2019] [Accepted: 04/30/2019] [Indexed: 12/23/2022]
Abstract
The genetic underpinnings of schizophrenia (SCZ) remain unclear. SCZ genetic studies thus far have only identified numerous single nucleotide polymorphisms with small effect sizes and a handful of copy number variants (CNVs). This study investigates the prevalence of well-characterized CNV syndromes and candidate CNVs within a cohort of 348 SCZ patients, and explores correlations to their phenotypic findings. There was an enrichment of syndromic CNVs in the cohort, as well as brain-related and immune pathway genes within the detected CNVs. SCZ patients with brain-related CNVs had increased CNV burden, neurodevelopmental features, and types of hallucinations. Based on these results, we propose a CNV-SCZ model wherein specific phenotypic profiles should be prioritized for CNV screening within the SCZ patient population.
Collapse
Affiliation(s)
- Venuja Sriretnakumar
- Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, 250 College Street, Toronto M5T 1R8, Canada
| | - Clement C Zai
- Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, 250 College Street, Toronto M5T 1R8, Canada
| | - Syed Wasim
- The Fred A. Litwin Family Centre in Genetic Medicine, University Health Network & Mount Sinai Hospital, 60 Murray Street, Toronto M5T 3L9, Canada
| | - Brianna Barsanti-Innes
- Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, 250 College Street, Toronto M5T 1R8, Canada
| | - James L Kennedy
- Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, 250 College Street, Toronto M5T 1R8, Canada
| | - Joyce So
- Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, 250 College Street, Toronto M5T 1R8, Canada; The Fred A. Litwin Family Centre in Genetic Medicine, University Health Network & Mount Sinai Hospital, 60 Murray Street, Toronto M5T 3L9, Canada.
| |
Collapse
|
34
|
Taoufik E, Kouroupi G, Zygogianni O, Matsas R. Synaptic dysfunction in neurodegenerative and neurodevelopmental diseases: an overview of induced pluripotent stem-cell-based disease models. Open Biol 2019; 8:rsob.180138. [PMID: 30185603 PMCID: PMC6170506 DOI: 10.1098/rsob.180138] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 08/14/2018] [Indexed: 12/12/2022] Open
Abstract
Synaptic dysfunction in CNS disorders is the outcome of perturbations in physiological synapse structure and function, and can be either the cause or the consequence in specific pathologies. Accumulating data in the field of neuropsychiatric disorders, including autism spectrum disorders, schizophrenia and bipolar disorder, point to a neurodevelopmental origin of these pathologies. Due to a relatively early onset of behavioural and cognitive symptoms, it is generally acknowledged that mental illness initiates at the synapse level. On the other hand, synaptic dysfunction has been considered as an endpoint incident in neurodegenerative diseases, such as Alzheimer's, Parkinson's and Huntington's, mainly due to the considerably later onset of clinical symptoms and progressive appearance of cognitive deficits. This dichotomy has recently been challenged, particularly since the discovery of cell reprogramming technologies and the generation of induced pluripotent stem cells from patient somatic cells. The creation of 'disease-in-a-dish' models for multiple CNS pathologies has revealed unexpected commonalities in the molecular and cellular mechanisms operating in both developmental and degenerative conditions, most of which meet at the synapse level. In this review we discuss synaptic dysfunction in prototype neurodevelopmental and neurodegenerative diseases, emphasizing overlapping features of synaptopathy that have been suggested by studies using induced pluripotent stem-cell-based systems. These valuable disease models have highlighted a potential neurodevelopmental component in classical neurodegenerative diseases that is worth pursuing and investigating further. Moving from demonstration of correlation to understanding mechanistic causality forms the basis for developing novel therapeutics.
Collapse
Affiliation(s)
- Era Taoufik
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, 127 Vassilissis Sofias Avenue, 11521 Athens, Greece
| | - Georgia Kouroupi
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, 127 Vassilissis Sofias Avenue, 11521 Athens, Greece
| | - Ourania Zygogianni
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, 127 Vassilissis Sofias Avenue, 11521 Athens, Greece
| | - Rebecca Matsas
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, 127 Vassilissis Sofias Avenue, 11521 Athens, Greece
| |
Collapse
|
35
|
Forsingdal A, Jørgensen TN, Olsen L, Werge T, Didriksen M, Nielsen J. Can Animal Models of Copy Number Variants That Predispose to Schizophrenia Elucidate Underlying Biology? Biol Psychiatry 2019; 85:13-24. [PMID: 30144930 DOI: 10.1016/j.biopsych.2018.07.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 06/15/2018] [Accepted: 07/03/2018] [Indexed: 12/21/2022]
Abstract
The diagnosis of schizophrenia rests on clinical criteria that cannot be assessed in animal models. Together with absence of a clear underlying pathology and understanding of what causes schizophrenia, this has hindered development of informative animal models. However, recent large-scale genomic studies have identified copy number variants (CNVs) that confer high risk of schizophrenia and have opened a new avenue for generation of relevant animal models. Eight recurrent CNVs have reproducibly been shown to increase the risk of schizophrenia by severalfold: 22q11.2(del), 15q13.3(del), 1q21(del), 1q21(dup), NRXN1(del), 3q29(del), 7q11.23(dup), and 16p11.2(dup). Five of these CNVs have been modeled in animals, mainly mice, but also rats, flies, and zebrafish, and have been shown to recapitulate behavioral and electrophysiological aspects of schizophrenia. Here, we provide an overview of the schizophrenia-related phenotypes found in animal models of schizophrenia high-risk CNVs. We also discuss strengths and limitations of the CNV models, and how they can advance our biological understanding of mechanisms that can lead to schizophrenia and can be used to develop new and better treatments for schizophrenia.
Collapse
Affiliation(s)
- Annika Forsingdal
- Division of Synaptic Transmission, H. Lundbeck A/S, Valby, Mental Health Center, Sankt Hans Hospital, Mental Health Services, Roskilde; Institute of Biological Psychiatry, Mental Health Center, Sankt Hans Hospital, Mental Health Services, Roskilde; Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
| | - Trine Nygaard Jørgensen
- Division of Synaptic Transmission, H. Lundbeck A/S, Valby, Mental Health Center, Sankt Hans Hospital, Mental Health Services, Roskilde
| | - Line Olsen
- Institute of Biological Psychiatry, Mental Health Center, Sankt Hans Hospital, Mental Health Services, Roskilde; iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
| | - Thomas Werge
- Institute of Biological Psychiatry, Mental Health Center, Sankt Hans Hospital, Mental Health Services, Roskilde; Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark; iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
| | - Michael Didriksen
- Division of Synaptic Transmission, H. Lundbeck A/S, Valby, Mental Health Center, Sankt Hans Hospital, Mental Health Services, Roskilde
| | - Jacob Nielsen
- Division of Synaptic Transmission, H. Lundbeck A/S, Valby, Mental Health Center, Sankt Hans Hospital, Mental Health Services, Roskilde.
| |
Collapse
|
36
|
Piras IS, Manchia M, Huentelman MJ, Pinna F, Zai CC, Kennedy JL, Carpiniello B. Peripheral Biomarkers in Schizophrenia: A Meta-Analysis of Microarray Gene Expression Datasets. Int J Neuropsychopharmacol 2018; 22:186-193. [PMID: 30576541 PMCID: PMC6403089 DOI: 10.1093/ijnp/pyy103] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 12/07/2018] [Accepted: 12/19/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Schizophrenia is a severe psychiatric disorder with a complex pathophysiology. Given its prevalence, high risk of mortality, early onset, and high levels of disability, researchers have attempted to develop early detection strategies for facilitating timely pharmacological and/or nonpharmacological interventions. Here, we performed a meta-analysis of publicly available gene expression datasets in peripheral tissues in schizophrenia and healthy controls to detect consistent patterns of illness-associated gene expression. We also tested whether our earlier finding of a downregulation of NPTX2 expression in the brain of schizophrenia patients replicated in peripheral tissues. METHODS We conducted a systematic search in the Gene Expression Omnibus repository (https://www.ncbi.nlm.nih.gov/gds/) and identified 3 datasets matching our inclusion criteria: GSE62333, GSE18312, and GSE27383. After quality controls, the total sample size was: schizophrenia (n = 71) and healthy controls (n = 57) (schizophrenia range: n = 12-40; healthy controls range: n = 8-29). RESULTS The results of the meta-analysis conducted with the GeneMeta package revealed 2 genes with a false discovery rate < 0.05: atlastin GTPase 3 (ATL3) (upregulated) and arachidonate 15-lipoxygenase, type B (ALOX15B) (downregulated). The result for ATL3 was confirmed using the weighted Z test method, whereas we found a suggestive signal for ALOX15B (false discovery rate < 0.10). CONCLUSIONS These data point to alterations of peripheral expression of ATL3 in schizophrenia, but did not confirm the significant association signal found for NPTX2 in postmortem brain samples. These findings await replication in newly recruited schizophrenia samples as well as complementary analysis of their encoded peptides in blood.
Collapse
Affiliation(s)
- Ignazio S Piras
- Neurogenomic Division, Translational Genomic Research Institute, Phoenix, Arizona
| | - Mirko Manchia
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy,Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada,Correspondence: Mirko Manchia, MD, PhD, Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Via Liguria, 13 - 09127 Cagliari, Italy ( and )
| | - Matthew J Huentelman
- Neurogenomic Division, Translational Genomic Research Institute, Phoenix, Arizona
| | - Federica Pinna
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Clement C Zai
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada,Department of Psychiatry, Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada,Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - James L Kennedy
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada,Department of Psychiatry, Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada,Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Bernardo Carpiniello
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
37
|
Yin L, Cheung EFC, Chen RYL, Wong EHM, Sham PC, So HC. Leveraging genome-wide association and clinical data in revealing schizophrenia subgroups. J Psychiatr Res 2018; 106:106-117. [PMID: 30312963 DOI: 10.1016/j.jpsychires.2018.09.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 09/13/2018] [Accepted: 09/18/2018] [Indexed: 02/04/2023]
Abstract
Schizophrenia (SCZ) has long been recognized as a highly heterogeneous disorder. Patients differed in their clinical manifestations, prognosis, and underlying pathophysiologies. Here we presented and applied a framework for finding subtypes of SCZ utilizing genome-wide association study (GWAS) and clinical data. We postulated that genetic information may help stratify patient into useful subgroups, and incorporation of other clinical information and cognitive profiles will further improve patient subtyping. We conducted cluster analysis in 387 Hong Kong Chinese with SCZ. First we performed 'single-view' clustering using genetic or clinical data alone, then proceeded to 'multi-view' clustering (MVC) accounting for both types of information. We validated clustering results by assessing subgroup differences in various outcomes. We found significant differences in outcomes including treatment response, disease course and symptom severity (Simes overall p-value using MVC = 1.64E-9). Overall speaking, we identified three subgroups with good, intermediate and poor prognosis respectively. MVC generally out-performed single-view methods. The analysis was repeated for different sets of input SNPs, and stratified analysis of male and female patients, and the results remained largely robust. We also found significant enrichment for SCZ loci among the SNPs selected by the cluster algorithm. Numerous selected genes (e.g. NRG1, ERBB4, NRXN1, ANK3) and pathways (e.g. neuregulin-ErbB4 and calcium signaling) were implicated in SCZ or related pathophysiological processes. This is first study to combine both genetic and clinical data for subtyping SCZ, and to employ genome-wide SNP data in cluster analysis of a complex disease. This work points to a new way of GWAS analysis of translational potential.
Collapse
Affiliation(s)
- Liangying Yin
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Eric Fuk-Chi Cheung
- Castle Peak Hospital, Hong Kong; Department of Psychiatry, University of Hong Kong, Hong Kong
| | | | | | - Pak-Chung Sham
- Department of Psychiatry, University of Hong Kong, Hong Kong; Centre for Genomic Sciences, University of Hong Kong, Hong Kong; State Key Laboratory for Cognitive and Brain Sciences, University of Hong Kong, Hong Kong
| | - Hon-Cheong So
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong; KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Zoology Institute of Zoology and the Chinese University of Hong Kong, China.
| |
Collapse
|
38
|
Saint-Martin M, Joubert B, Pellier-Monnin V, Pascual O, Noraz N, Honnorat J. Contactin-associated protein-like 2, a protein of the neurexin family involved in several human diseases. Eur J Neurosci 2018; 48:1906-1923. [PMID: 30028556 DOI: 10.1111/ejn.14081] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/08/2018] [Accepted: 07/02/2018] [Indexed: 12/11/2022]
Abstract
Contactin-associated protein-like 2 (CASPR2) is a cell adhesion protein of the neurexin family. Proteins of this family have been shown to play a role in the development of the nervous system, in synaptic functions, and in neurological diseases. Over recent years, CASPR2 function has gained an increasing interest as demonstrated by the growing number of publications. Here, we gather published data to comprehensively review CASPR2 functions within the nervous system in relation to CASPR2-related diseases in humans. On the one hand, studies on Cntnap2 (coding for CASPR2) knockout mice revealed its role during development, especially, in setting-up the inhibitory network. Consistent with this result, mutations in the CNTNAP2 gene coding for CASPR2 in human have been identified in neurodevelopmental disorders such as autism, intellectual disability, and epilepsy. On the other hand, CASPR2 was shown to play a role beyond development, in the localization of voltage-gated potassium channel (VGKC) complex that is composed of TAG-1, Kv1.1, and Kv1.2. This complex was found in several subcellular compartments essential for action potential propagation: the node of Ranvier, the axon initial segment, and the synapse. In line with a role of CASPR2 in the mature nervous system, neurological autoimmune diseases have been described in patients without neurodevelopmental disorders but with antibodies directed against CASPR2. These autoimmune diseases were of two types: central with memory disorders and temporal lobe seizures, or peripheral with muscular hyperactivity. Overall, we review the up-to-date knowledge on CASPR2 function and pinpoint confused or lacking information that will need further investigation.
Collapse
Affiliation(s)
- Margaux Saint-Martin
- Institut NeuroMyoGene INSERM U1217/CNRS UMR 5310, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Bastien Joubert
- Institut NeuroMyoGene INSERM U1217/CNRS UMR 5310, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France.,French Reference Center on Paraneoplastic Neurological Syndrome, Hospices Civils de Lyon, Hôpital Neurologique, Bron, France
| | - Véronique Pellier-Monnin
- Institut NeuroMyoGene INSERM U1217/CNRS UMR 5310, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Olivier Pascual
- Institut NeuroMyoGene INSERM U1217/CNRS UMR 5310, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Nelly Noraz
- Institut NeuroMyoGene INSERM U1217/CNRS UMR 5310, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Jérôme Honnorat
- Institut NeuroMyoGene INSERM U1217/CNRS UMR 5310, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France.,French Reference Center on Paraneoplastic Neurological Syndrome, Hospices Civils de Lyon, Hôpital Neurologique, Bron, France
| |
Collapse
|
39
|
Sullivan CR, O'Donovan SM, McCullumsmith RE, Ramsey A. Defects in Bioenergetic Coupling in Schizophrenia. Biol Psychiatry 2018; 83:739-750. [PMID: 29217297 PMCID: PMC5891385 DOI: 10.1016/j.biopsych.2017.10.014] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/18/2017] [Accepted: 10/09/2017] [Indexed: 02/06/2023]
Abstract
Synaptic neurotransmission relies on maintenance of the synapse and meeting the energy demands of neurons. Defects in excitatory and inhibitory synapses have been implicated in schizophrenia, likely contributing to positive and negative symptoms as well as impaired cognition. Recently, accumulating evidence has suggested that bioenergetic systems, important in both synaptic function and cognition, are abnormal in psychiatric illnesses such as schizophrenia. Animal models of synaptic dysfunction demonstrated endophenotypes of schizophrenia as well as bioenergetic abnormalities. We report findings on the bioenergetic interplay of astrocytes and neurons and discuss how dysregulation of these pathways may contribute to the pathogenesis of schizophrenia, highlighting metabolic systems as important therapeutic targets.
Collapse
Affiliation(s)
- Courtney R Sullivan
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, Ohio
| | - Sinead M O'Donovan
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, Ohio
| | - Robert E McCullumsmith
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, Ohio.
| | - Amy Ramsey
- Department of Pharmacology and Toxicology, University of Toronto, Ontario, Canada
| |
Collapse
|
40
|
Giannitelli M, Consoli A, Raffin M, Jardri R, Levinson DF, Cohen D, Laurent-Levinson C. An overview of medical risk factors for childhood psychosis: Implications for research and treatment. Schizophr Res 2018; 192:39-49. [PMID: 28526280 DOI: 10.1016/j.schres.2017.05.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 05/05/2017] [Accepted: 05/08/2017] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Psychotic disorders in childhood and early adolescence often progress to chronic schizophrenia, but in many cases there are diagnosable medical and genetic causes or risk factors. We reviewed our clinical experience and the relevant literature to identify these factors and to define their clinical features, appropriate work-up and treatment. METHOD We reviewed the results of comprehensive medical evaluations of 160 psychotic children and adolescents in our center. We also searched the Medline database (January 1994 to December 2015) with the following keywords and combinations: early onset schizophrenia, childhood onset schizophrenia, early onset psychosis, first episode psychosis, inborn errors of metabolism (IEM), genetic syndrome, copy number variants, autoimmune disorders, endocrine diseases, nutritional deficiencies, central nervous system infections, movement disorders, and epilepsy. RESULTS In our center, 12.5% of cases had medical disorders likely to be contributing to psychosis. Based on 66 relevant papers and our experience, we describe the clinical features of multiple genetic syndromes, IEM, and autoimmune, neurological, endocrinological and nutritional disorders that increase the risk of psychotic disorders in childhood and adolescence. We propose an algorithm for systematic laboratory evaluation, informed by clinical examination, emphasizing common and/or treatable factors. CONCLUSIONS In children and early adolescents with psychotic disorders, systematic medical work-up is warranted to identify medical and genetic factors. Not every rare cause can be worked up, thus careful clinical examinations are required to detect medical, neurological and genetic signs. Comprehensive medical evaluation can detect treatable diseases among cases of early-onset psychosis.
Collapse
Affiliation(s)
- Marianna Giannitelli
- Sorbonne Universités, UPMC Univ Paris 06, Assistance Publique-Hôpitaux de Paris, Groupe de Recherche Clinique n°15 (PSYDEV), Hôpital Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, 75013 Paris, France; Centre de référence des maladies rares à expression psychiatrique, Department of Child and Adolescent Psychiatry, Hôpital Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, 75013 Paris, France; CNRS UMR 7222, Institut des Systèmes Intelligents et Robotiques, Université Pierre et Marie Curie, 1 place Jussieu, 75005 Paris, France
| | - Angèle Consoli
- Sorbonne Universités, UPMC Univ Paris 06, Assistance Publique-Hôpitaux de Paris, Groupe de Recherche Clinique n°15 (PSYDEV), Hôpital Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, 75013 Paris, France; Centre de référence des maladies rares à expression psychiatrique, Department of Child and Adolescent Psychiatry, Hôpital Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, 75013 Paris, France
| | - Marie Raffin
- Sorbonne Universités, UPMC Univ Paris 06, Assistance Publique-Hôpitaux de Paris, Groupe de Recherche Clinique n°15 (PSYDEV), Hôpital Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, 75013 Paris, France
| | - Renaud Jardri
- University of Lille, SCALab, CNRS UMR-9193 & CHU Lille, CURE platform, Fontan Hospital, Lille, France
| | - Douglas F Levinson
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - David Cohen
- Sorbonne Universités, UPMC Univ Paris 06, Assistance Publique-Hôpitaux de Paris, Groupe de Recherche Clinique n°15 (PSYDEV), Hôpital Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, 75013 Paris, France; Centre de référence des maladies rares à expression psychiatrique, Department of Child and Adolescent Psychiatry, Hôpital Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, 75013 Paris, France; CNRS UMR 7222, Institut des Systèmes Intelligents et Robotiques, Université Pierre et Marie Curie, 1 place Jussieu, 75005 Paris, France
| | - Claudine Laurent-Levinson
- Sorbonne Universités, UPMC Univ Paris 06, Assistance Publique-Hôpitaux de Paris, Groupe de Recherche Clinique n°15 (PSYDEV), Hôpital Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, 75013 Paris, France; Centre de référence des maladies rares à expression psychiatrique, Department of Child and Adolescent Psychiatry, Hôpital Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, 75013 Paris, France; Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
| |
Collapse
|
41
|
Selten M, van Bokhoven H, Nadif Kasri N. Inhibitory control of the excitatory/inhibitory balance in psychiatric disorders. F1000Res 2018; 7:23. [PMID: 29375819 PMCID: PMC5760969 DOI: 10.12688/f1000research.12155.1] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/21/2017] [Indexed: 12/21/2022] Open
Abstract
Neuronal networks consist of different types of neurons that all play their own role in order to maintain proper network function. The two main types of neurons segregate in excitatory and inhibitory neurons, which together regulate the flow of information through the network. It has been proposed that changes in the relative strength in these two opposing forces underlie the symptoms observed in psychiatric disorders, including autism and schizophrenia. Here, we review the role of alterations to the function of the inhibitory system as a cause of psychiatric disorders. First, we explore both patient and post-mortem evidence of inhibitory deficiency. We then discuss the function of different interneuron subtypes in the network and focus on the central role of a specific class of inhibitory neurons, parvalbumin-positive interneurons. Finally, we discuss genes known to be affected in different disorders and the effects that mutations in these genes have on the inhibitory system in cortex and hippocampus. We conclude that alterations to the inhibitory system are consistently identified in animal models of psychiatric disorders and, more specifically, that mutations affecting the function of parvalbumin-positive interneurons seem to play a central role in the symptoms observed in these disorders.
Collapse
Affiliation(s)
- Martijn Selten
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, New Hunt's House, Guy's Campus, King's College London, London, SE1 1UL, UK.,MRC Centre for Neurodevelopmental Disorders, New Hunt's House, Guy's Campus, King's College London, London, SE1 1UL, UK.,Department of Human Genetics & Department of Cognitive Neuroscience, Radboudumc, Geert Grooteplein 10, Box 9101, 6500 HB Nijmegen, Netherlands.,Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ Nijmegen, Netherlands
| | - Hans van Bokhoven
- Department of Human Genetics & Department of Cognitive Neuroscience, Radboudumc, Geert Grooteplein 10, Box 9101, 6500 HB Nijmegen, Netherlands.,Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ Nijmegen, Netherlands
| | - Nael Nadif Kasri
- Department of Human Genetics & Department of Cognitive Neuroscience, Radboudumc, Geert Grooteplein 10, Box 9101, 6500 HB Nijmegen, Netherlands.,Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ Nijmegen, Netherlands
| |
Collapse
|
42
|
Glessner JT, Li J, Wang D, March M, Lima L, Desai A, Hadley D, Kao C, Gur RE, Cohen N, Sleiman PMA, Li Q, Hakonarson H. Copy number variation meta-analysis reveals a novel duplication at 9p24 associated with multiple neurodevelopmental disorders. Genome Med 2017; 9:106. [PMID: 29191242 PMCID: PMC5709845 DOI: 10.1186/s13073-017-0494-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/14/2017] [Indexed: 12/24/2022] Open
Abstract
Background Neurodevelopmental and neuropsychiatric disorders represent a wide spectrum of heterogeneous yet inter-related disease conditions. The overlapping clinical presentations of these diseases suggest a shared genetic etiology. We aim to identify shared structural variants spanning the spectrum of five neuropsychiatric disorders. Methods We investigated copy number variations (CNVs) in five cohorts, including schizophrenia (SCZ), bipolar disease (BD), autism spectrum disorders (ASD), attention deficit hyperactivity disorder (ADHD), and depression, from 7849 cases and 10,799 controls. CNVs were called based on intensity data from genome-wide SNP arrays and CNV frequency was compared between cases and controls in each disease cohort separately. Meta-analysis was performed via a gene-based approach. Quantitative PCR (qPCR) was employed to validate novel significant loci. Results In our meta-analysis, two genes containing CNVs with exonic overlap reached genome-wide significance threshold of meta P value < 9.4 × 10−6 for deletions and 7.5 × 10−6 for duplications. We observed significant overlap between risk CNV loci across cohorts. In addition, we identified novel significant associations of DOCK8/KANK1 duplications (meta P value = 7.5 × 10−7) across all cohorts, and further validated the CNV region with qPCR. Conclusions In the first large scale meta-analysis of CNVs across multiple neurodevelopmental/psychiatric diseases, we uncovered novel significant associations of structural variants in the locus of DOCK8/KANK1 shared by five diseases, suggesting common etiology of these clinically distinct neurodevelopmental conditions. Electronic supplementary material The online version of this article (doi:10.1186/s13073-017-0494-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Joseph T Glessner
- The Center for Applied Genomics, Abramson Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Jin Li
- The Center for Applied Genomics, Abramson Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.,Department of Cell Biology, Tianjin Medical University, Tianjin, China
| | - Dai Wang
- Janssen Research & Development, LLC, Raritan, NJ, 08869, USA
| | - Michael March
- The Center for Applied Genomics, Abramson Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Leandro Lima
- The Center for Applied Genomics, Abramson Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Akshatha Desai
- The Center for Applied Genomics, Abramson Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Dexter Hadley
- The Center for Applied Genomics, Abramson Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Charlly Kao
- The Center for Applied Genomics, Abramson Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Raquel E Gur
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Nadine Cohen
- Janssen Research & Development, LLC, Raritan, NJ, 08869, USA
| | - Patrick M A Sleiman
- The Center for Applied Genomics, Abramson Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Qingqin Li
- Janssen Research & Development, LLC, Titusville, NJ, 08560, USA
| | - Hakon Hakonarson
- The Center for Applied Genomics, Abramson Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA. .,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA. .,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.
| | | |
Collapse
|
43
|
Giegling I, Hosak L, Mössner R, Serretti A, Bellivier F, Claes S, Collier DA, Corrales A, DeLisi LE, Gallo C, Gill M, Kennedy JL, Leboyer M, Maier W, Marquez M, Massat I, Mors O, Muglia P, Nöthen MM, Ospina-Duque J, Owen MJ, Propping P, Shi Y, St Clair D, Thibaut F, Cichon S, Mendlewicz J, O'Donovan MC, Rujescu D. Genetics of schizophrenia: A consensus paper of the WFSBP Task Force on Genetics. World J Biol Psychiatry 2017; 18:492-505. [PMID: 28112043 DOI: 10.1080/15622975.2016.1268715] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 11/29/2016] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Schizophrenia is a severe psychiatric disease affecting about 1% of the general population. The relative contribution of genetic factors has been estimated to be up to 80%. The mode of inheritance is complex, non-Mendelian, and in most cases involving the combined action of large numbers of genes. METHODS This review summarises recent efforts to identify genetic variants associated with schizophrenia detected, e.g., through genome-wide association studies, studies on copy-number variants or next-generation sequencing. RESULTS A large, new body of evidence on genetics of schizophrenia has accumulated over recent years. Many new robustly associated genetic loci have been detected. Furthermore, there is consensus that at least a dozen microdeletions and microduplications contribute to the disease. Genetic overlap between schizophrenia, other psychiatric disorders, and neurodevelopmental syndromes raised new questions regarding the current classification of psychiatric and neurodevelopmental diseases. CONCLUSIONS Future studies will address especially the functional characterisation of genetic variants. This will hopefully open the doors to our understanding of the pathophysiology of schizophrenia and other related diseases. Complementary, integrated systems biology approaches to genomics, transcriptomics, proteomics and metabolomics may also play crucial roles in enabling a precision medicine approach to the treatment of individual patients.
Collapse
Affiliation(s)
- Ina Giegling
- a Department of Psychiatry, Psychotherapy, and Psychosomatics , Martin Luther University of Halle-Wittenberg , Halle , Germany
- b Department of Psychiatry , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Ladislav Hosak
- c Department of Psychiatriy , Charles University, Faculty of Medicine and University Hospital in Hradec Králové, Prague , Czech Republic
| | - Rainald Mössner
- d Department of Psychiatry and Psychotherapy , University of Tübingen , Tübingen , Germany
| | - Alessandro Serretti
- e Department of Biomedical and Neuromotor Sciences , University of Bologna , Bologna , Italy
| | - Frank Bellivier
- f Fondation Fondamental, Créteil, France AP-HP, GH Saint-Louis-Lariboisière-Fernand-Widal, Pôle Neurosciences , Paris , France
- g Equipe 1, Université Paris Diderot , Paris , France
| | - Stephan Claes
- h GRASP-Research Group, Department of Neuroscience , University of Leuven , Leuven , Belgium
- i Department of Neurosciences, University Psychiatric Center KU Leuven , Leuven , Belgium
| | - David A Collier
- j Social, Genetic and Developmental Psychiatry Centre , Institute of Psychiatry, King's College London , London , UK
- k Eli Lilly and Company Ltd, Erl Wood Manor , Surrey , UK
| | - Alejo Corrales
- l Argentinean Association of Biological Psychiatry , National University, UNT, Buenos Aires , Argentina
| | - Lynn E DeLisi
- m VA Boston Health Care System , Brockton , MA , USA
- n Department of Psychiatry , Harvard Medical School , Boston , MA , USA
| | - Carla Gallo
- o Departamento de Ciencias Celulares y Moleculares, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía , Universidad Peruana Cayetano Heredia , Lima , Peru
| | - Michael Gill
- p Neuropsychiatric Genetics Research Group, Department of Psychiatry , Trinity College Dublin , Dublin , Ireland
| | - James L Kennedy
- q Neurogenetics Section, Centre for Addiction and Mental Health , Toronto , ON , Canada
- r Centre for Addiction and Mental Health , Campbell Family Mental Health Research Institute , Toronto , ON , Canada
- s Department of Psychiatry , University of Toronto , Toronto , ON , Canada
- t Collaborative Program in Neuroscience, Institute of Medical Science, University of Toronto , Toronto , ON , Canada
| | - Marion Leboyer
- u Equipe Psychiatrie Translationnelle, Faculté de Médecine, Université Paris-Est Créteil, Inserm U955 , Créteil , France
- v DHU Pe-Psy, Pôle de Psychiatrie et d'Addictologie , AP-HP, Hôpitaux Universitaires Henri Mondor , Créteil , France
- w Pôle de Psychiatrie , Hôpital Albert Chenevier , Créteil , France
- x Fondation FondaMental , Créteil , France
| | - Wolfgang Maier
- y Department of Psychiatry and Psychotherapy , University of Bonn, Bonn , Germany
| | - Miguel Marquez
- z Asistencia, Docencia e Investigación en Neurociencia , Buenos Aires , Argentina
| | - Isabelle Massat
- aa UNI - ULB Neurosciences Institute, ULB , Bruxelles , Belgium
- ab National Fund of Scientific Research (FNRS) , Bruxelles , Belgium
- ac Laboratory of Experimental Neurology , ULB , Bruxelles , Belgium
- ad UR2NF - Neuropsychology and Functional Neuroimaging Research Unit, Centre de Recherche Cognition et Neurosciences , Université Libre de Bruxelles (ULB) , Bruxelles , Belgium
| | - Ole Mors
- ae Psychosis Research Unit , Aarhus University Hospital , Risskov , Denmark
- af The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus , Denmark
| | | | - Markus M Nöthen
- ah Head, Institute of Human Genetics, University of Bonn , Bonn , Germany
- ai Department of Genomics , Life and Brain Center , Bonn , Germany
| | - Jorge Ospina-Duque
- aj Grupo de Investigación en Psiquiatría, Departamento de Psiquiatría, Facultad de Medicina , Universidad de Antioquia , Medellín , Colombia
| | - Michael J Owen
- ak MRC Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, School of Medicine , Cardiff University , Cardiff , UK
- al National Centre for Mental Health, Cardiff University , Cardiff , UK
| | | | - YongYong Shi
- an Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education , Shanghai Jiao Tong University , Shanghai , China
- ao Shandong Provincial Key Laboratory of Metabloic Disease, The Affiliated Hospital of Qingdao University , Qingdao , P.R. China
- ap Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University , Shanghai , P.R. China
| | - David St Clair
- aq Department of Psychiatry, University of Aberdeen, Institute of Medical Sciences , Aberdeen , UK
| | - Florence Thibaut
- ar INSERM U 894 Centre Psychiatry and Neurosciences , University Hospital Cochin (Site Tarnier), University Sorbonne Paris Cité (Faculty of Medicine Paris Descartes) , Paris , France
| | - Sven Cichon
- ah Head, Institute of Human Genetics, University of Bonn , Bonn , Germany
- ai Department of Genomics , Life and Brain Center , Bonn , Germany
- as Division of Medical Genetics, Department of Biomedicine , University of Basel , Basel , Switzerland
- at Genomic Imaging, Institute of Neuroscience and Medicine , Research Center Juelich , Juelich , Germany
| | - Julien Mendlewicz
- au Laboratoire de Psychologie Medicale, Centre Europe´en de Psychologie Medicale , Universite´ Libre de Bruxelles and Psy Pluriel , Brussels , Belgium
| | - Michael C O'Donovan
- ak MRC Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, School of Medicine , Cardiff University , Cardiff , UK
- al National Centre for Mental Health, Cardiff University , Cardiff , UK
| | - Dan Rujescu
- a Department of Psychiatry, Psychotherapy, and Psychosomatics , Martin Luther University of Halle-Wittenberg , Halle , Germany
- b Department of Psychiatry , Ludwig-Maximilians-University Munich , Munich , Germany
| |
Collapse
|
44
|
Nowakowska B. Clinical interpretation of copy number variants in the human genome. J Appl Genet 2017; 58:449-457. [PMID: 28963714 PMCID: PMC5655614 DOI: 10.1007/s13353-017-0407-4] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 08/31/2017] [Accepted: 09/05/2017] [Indexed: 12/15/2022]
Abstract
Molecular methods, by which copy number variants (CNVs) detection is available, have been gradually introduced into routine diagnostics over the last 15 years. Despite this, some CNVs continue to be a huge challenge when it comes to clinical interpretation. CNVs are an important source of normal and pathogenic variants, but, in many cases, their impact on human health depends on factors that are not yet known. Therefore, perception of their clinical consequences can change over time, as our knowledge grows. This review summarises guidelines that facilitate correct classification of identified changes and discusses difficulties with the interpretation of rare, small CNVs.
Collapse
Affiliation(s)
- Beata Nowakowska
- Department of Medical Genetics, Institute of Mother and Child, Kasprzaka 17a, 01-211, Warsaw, Poland.
| |
Collapse
|
45
|
Song W, Tavitian A, Cressatti M, Galindez C, Liberman A, Schipper HM. Cysteine-rich whey protein isolate (Immunocal®) ameliorates deficits in the GFAP.HMOX1 mouse model of schizophrenia. Free Radic Biol Med 2017; 110:162-175. [PMID: 28603087 DOI: 10.1016/j.freeradbiomed.2017.05.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 05/26/2017] [Accepted: 05/30/2017] [Indexed: 12/22/2022]
Abstract
Schizophrenia is a neuropsychiatric disorder that features neural oxidative stress and glutathione (GSH) deficits. Oxidative stress is augmented in brain tissue of GFAP.HMOX1 transgenic mice which exhibit schizophrenia-relevant characteristics. The whey protein isolate, Immunocal® serves as a GSH precursor upon oral administration. In this study, we treated GFAP.HMOX1 transgenic mice daily with either Immunocal (33mg/ml drinking water) or equivalent concentrations of casein (control) between the ages of 5 and 6.5 months. Immunocal attenuated many of the behavioral, neurochemical and redox abnormalities observed in GFAP.HMOX1 mice. In addition to restoring GSH homeostasis in the CNS of the transgenic mice, the whey protein isolate augmented GSH reserves in the brains of wild-type animals. These results demonstrate that consumption of whey protein isolate augments GSH stores and antioxidant defenses in the healthy and diseased mammalian brain. Whey protein isolate supplementation (Immunocal) may constitute a safe and effective modality for the management of schizophrenia, an unmet clinical imperative.
Collapse
Affiliation(s)
- Wei Song
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3999 Cote Ste. Catherine Road, Montreal, Quebec, Canada H3T 1E2.
| | - Ayda Tavitian
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3999 Cote Ste. Catherine Road, Montreal, Quebec, Canada H3T 1E2; Department of Neurology and Neurosurgery, McGill University, 3801 University Street, Montreal, Quebec, Canada H3A 2B4.
| | - Marisa Cressatti
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3999 Cote Ste. Catherine Road, Montreal, Quebec, Canada H3T 1E2; Department of Neurology and Neurosurgery, McGill University, 3801 University Street, Montreal, Quebec, Canada H3A 2B4.
| | - Carmela Galindez
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3999 Cote Ste. Catherine Road, Montreal, Quebec, Canada H3T 1E2.
| | - Adrienne Liberman
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3999 Cote Ste. Catherine Road, Montreal, Quebec, Canada H3T 1E2.
| | - Hyman M Schipper
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3999 Cote Ste. Catherine Road, Montreal, Quebec, Canada H3T 1E2; Department of Neurology and Neurosurgery, McGill University, 3801 University Street, Montreal, Quebec, Canada H3A 2B4.
| |
Collapse
|
46
|
Henriksen MG, Nordgaard J, Jansson LB. Genetics of Schizophrenia: Overview of Methods, Findings and Limitations. Front Hum Neurosci 2017; 11:322. [PMID: 28690503 PMCID: PMC5480258 DOI: 10.3389/fnhum.2017.00322] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 06/06/2017] [Indexed: 01/12/2023] Open
Abstract
Genetics constitute a crucial risk factor to schizophrenia. In the last decade, molecular genetic research has produced novel findings, infusing optimism about discovering the biological roots of schizophrenia. However, the complexity of the object of inquiry makes it almost impossible for non-specialists in genetics (e.g., many clinicians and researchers) to get a proper understanding and appreciation of the genetic findings and their limitations. This study aims at facilitating such an understanding by providing a brief overview of some of the central methods and findings in schizophrenia genetics, from its historical origins to its current status, and also by addressing some limitations and challenges that confront this field of research. In short, the genetic architecture of schizophrenia has proven to be highly complex, heterogeneous and polygenic. The disease risk is constituted by numerous common genetic variants of only very small individual effect and by rare, highly penetrant genetic variants of larger effects. In spite of recent advances in molecular genetics, our knowledge of the etiopathogenesis of schizophrenia and the genotype-environment interactions remain limited.
Collapse
Affiliation(s)
- Mads G. Henriksen
- Mental Health Center Glostrup, University Hospital of CopenhagenCopenhagen, Denmark
- Faculty of Health and Medical Sciences, Institute of Clinical Medicine, University of CopenhagenCopenhagen, Denmark
- Center for Subjectivity Research, University of CopenhagenCopenhagen, Denmark
| | - Julie Nordgaard
- Faculty of Health and Medical Sciences, Institute of Clinical Medicine, University of CopenhagenCopenhagen, Denmark
- Early Psychosis Intervention Center, Region Zealand Psychiatry Roskilde, University of CopenhagenCopenhagen, Denmark
| | - Lennart B. Jansson
- Mental Health Center Glostrup, University Hospital of CopenhagenCopenhagen, Denmark
- Faculty of Health and Medical Sciences, Institute of Clinical Medicine, University of CopenhagenCopenhagen, Denmark
| |
Collapse
|
47
|
Zhuo C, Hou W, Lin C, Hu L, Li J. Potential Value of Genomic Copy Number Variations in Schizophrenia. Front Mol Neurosci 2017; 10:204. [PMID: 28680393 PMCID: PMC5478687 DOI: 10.3389/fnmol.2017.00204] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 06/09/2017] [Indexed: 11/13/2022] Open
Abstract
Schizophrenia is a devastating neuropsychiatric disorder affecting approximately 1% of the global population, and the disease has imposed a considerable burden on families and society. Although, the exact cause of schizophrenia remains unknown, several lines of scientific evidence have revealed that genetic variants are strongly correlated with the development and early onset of the disease. In fact, the heritability among patients suffering from schizophrenia is as high as 80%. Genomic copy number variations (CNVs) are one of the main forms of genomic variations, ubiquitously occurring in the human genome. An increasing number of studies have shown that CNVs account for population diversity and genetically related diseases, including schizophrenia. The last decade has witnessed rapid advances in the development of novel genomic technologies, which have led to the identification of schizophrenia-associated CNVs, insight into the roles of the affected genes in their intervals in schizophrenia, and successful manipulation of the target CNVs. In this review, we focus on the recent discoveries of important CNVs that are associated with schizophrenia and outline the potential values that the study of CNVs will bring to the areas of schizophrenia research, diagnosis, and therapy. Furthermore, with the help of the novel genetic tool known as the Clustered Regularly Interspaced Short Palindromic Repeats-associated nuclease 9 (CRISPR/Cas9) system, the pathogenic CNVs as genomic defects could be corrected. In conclusion, the recent novel findings of schizophrenia-associated CNVs offer an exciting opportunity for schizophrenia research to decipher the pathological mechanisms underlying the onset and development of schizophrenia as well as to provide potential clinical applications in genetic counseling, diagnosis, and therapy for this complex mental disease.
Collapse
Affiliation(s)
- Chuanjun Zhuo
- Department of Psychological Medicine, Wenzhou Seventh People's HospitalWenzhou, China.,Department of Psychological Medicine, Tianjin Anding HospitalTianjin, China
| | - Weihong Hou
- Department of Biology, University of North Carolina at CharlotteCharlotte, NC, United States.,Department of Biochemistry and Molecular Biology, Zhengzhou UniversityZhengzhou, China
| | - Chongguang Lin
- Department of Psychological Medicine, Wenzhou Seventh People's HospitalWenzhou, China
| | - Lirong Hu
- Department of Psychological Medicine, Wenzhou Seventh People's HospitalWenzhou, China
| | - Jie Li
- Department of Psychological Medicine, Tianjin Anding HospitalTianjin, China
| |
Collapse
|
48
|
|
49
|
Lanning R, Lett TA, Tiwari AK, Brandl EJ, de Luca V, Voineskos AN, Potkin SG, Lieberman JA, Meltzer HY, Müller DJ, Remington G, Kennedy JL, Zai CC. Association study between the neurexin-1 gene and tardive dyskinesia. Hum Psychopharmacol 2017; 32. [PMID: 28120489 DOI: 10.1002/hup.2568] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 10/16/2016] [Accepted: 11/28/2016] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Tardive dyskinesia (TD) is a motor side effect that may develop after long-term antipsychotic treatment. Schizophrenia has recently been associated with the Neurexin-1 (NRXN1) gene that codes for a cell adhesion molecule in synaptic communication. METHODS This study examined five NRXN1 single-nucleotide polymorphisms (SNPs) for possible association with the occurrence and severity of TD in 178 schizophrenia patients of European ancestry. RESULTS We did not find these SNPs to be significantly associated with TD. CONCLUSIONS More research is needed with additional SNPs and in bigger samples before we can completely rule out the role of NRXN1 in TD.
Collapse
Affiliation(s)
- Rachel Lanning
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Tristram A Lett
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry, Charité University Medicine Berlin, Berlin, Germany
| | - Arun K Tiwari
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Eva J Brandl
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry, Charité University Medicine Berlin, Berlin, Germany
| | - Vincenzo de Luca
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Aristotle N Voineskos
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Steven G Potkin
- Department of Psychiatry and Human Behavior, University of California, Irvine, California, USA
| | - Jeffrey A Lieberman
- Department of Psychiatry, Mental Health and Neuroscience Center, the University of North Carolina at Chapel Hill School of Medicine, North Carolina, USA
| | - Herbert Y Meltzer
- Psychiatry and Behavioral Sciences, Pharmacology and Physiology, Chemistry of Life Processes Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Daniel J Müller
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Gary Remington
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - James L Kennedy
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Clement C Zai
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.,Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
50
|
Nguyen TM, Schreiner D, Xiao L, Traunmüller L, Bornmann C, Scheiffele P. An alternative splicing switch shapes neurexin repertoires in principal neurons versus interneurons in the mouse hippocampus. eLife 2016; 5. [PMID: 27960072 PMCID: PMC5213383 DOI: 10.7554/elife.22757] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 12/07/2016] [Indexed: 01/18/2023] Open
Abstract
The unique anatomical and functional features of principal and interneuron populations are critical for the appropriate function of neuronal circuits. Cell type-specific properties are encoded by selective gene expression programs that shape molecular repertoires and synaptic protein complexes. However, the nature of such programs, particularly for post-transcriptional regulation at the level of alternative splicing is only beginning to emerge. We here demonstrate that transcripts encoding the synaptic adhesion molecules neurexin-1,2,3 are commonly expressed in principal cells and interneurons of the mouse hippocampus but undergo highly differential, cell type-specific alternative splicing. Principal cell-specific neurexin splice isoforms depend on the RNA-binding protein Slm2. By contrast, most parvalbumin-positive (PV+) interneurons lack Slm2, express a different neurexin splice isoform and co-express the corresponding splice isoform-specific neurexin ligand Cbln4. Conditional ablation of Nrxn alternative splice insertions selectively in PV+ cells results in elevated hippocampal network activity and impairment in a learning task. Thus, PV-cell-specific alternative splicing of neurexins is critical for neuronal circuit function DOI:http://dx.doi.org/10.7554/eLife.22757.001
Collapse
Affiliation(s)
| | | | - Le Xiao
- Biozentrum, University of Basel, Basel, Switzerland
| | | | | | | |
Collapse
|