151
|
den Hoed J, Fisher SE. Genetic pathways involved in human speech disorders. Curr Opin Genet Dev 2020; 65:103-111. [PMID: 32622339 DOI: 10.1016/j.gde.2020.05.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/08/2020] [Accepted: 05/08/2020] [Indexed: 12/12/2022]
Abstract
Rare genetic variants that disrupt speech development provide entry points for deciphering the neurobiological foundations of key human capacities. The value of this approach is illustrated by FOXP2, a transcription factor gene that was implicated in speech apraxia, and subsequently investigated using human cell-based systems and animal models. Advances in next-generation sequencing, coupled to de novo paradigms, facilitated discovery of etiological variants in additional genes in speech disorder cohorts. As for other neurodevelopmental syndromes, gene-driven studies show blurring of boundaries between diagnostic categories, with some risk genes shared across speech disorders, intellectual disability and autism. Convergent evidence hints at involvement of regulatory genes co-expressed in early human brain development, suggesting that etiological pathways could be amenable for investigation in emerging neural models such as cerebral organoids.
Collapse
Affiliation(s)
- Joery den Hoed
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, 6525 XD Nijmegen, The Netherlands; International Max Planck Research School for Language Sciences, Max Planck Institute for Psycholinguistics, 6525 XD Nijmegen, The Netherlands
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, 6525 XD Nijmegen, The Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 EN Nijmegen, The Netherlands.
| |
Collapse
|
152
|
Intermingling of gut microbiota with brain: Exploring the role of probiotics in battle against depressive disorders. Food Res Int 2020; 137:109489. [PMID: 33233143 DOI: 10.1016/j.foodres.2020.109489] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023]
Abstract
Depression is a debilitating psychiatric ailment which exerts disastrous effects on one's mental and physical health. Depression is accountable for augmentation of various life-threatening maladies such as neurodegenerative anomalies, cardiovascular diseases and diabetes. Depressive episodes are recurrent, pose a negative impact on life quality, decline life expectancy and enhance suicidal tendencies. Anti-depression chemotherapy displays marked adverse effects and frequent relapses. Thus, newer therapeutic interventions to prevent or combat depression are desperately required. Discovery of gut microbes as our mutualistic partner was made a long time ago and it is surprising that their functions still continue to expand and as of yet many are still to be uncovered. Experimental studies have revealed astonishing role of gut commensals in gut-brain signaling, immune homeostasis and hormonal regulation. Now, it is a well-established fact that gut microbes can alleviate stress or depression associated symptoms by modulating brain functions. Here in, we provide an overview of physiological alleyways involved in cross-talk between gut and brain, part played by probiotics in regulation of these pathways and use of probiotic bacteria as psychobiotics in various mental or depressive disorders.
Collapse
|
153
|
Roston A, Evans D, Gill H, McKinnon M, Isidor B, Cogné B, Mwenifumbo J, van Karnebeek C, An J, Jones SJM, Farrer M, Demos M, Connolly M, Gibson WT. SETD1B-associated neurodevelopmental disorder. J Med Genet 2020; 58:196-204. [PMID: 32546566 DOI: 10.1136/jmedgenet-2019-106756] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/11/2020] [Accepted: 04/14/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Dysfunction of histone methyltransferases and chromatin modifiers has been implicated in complex neurodevelopmental syndromes and cancers. SETD1B encodes a lysine-specific methyltransferase that assists in transcriptional activation of genes by depositing H3K4 methyl marks. Previous reports of patients with rare variants in SETD1B describe a distinctive phenotype that includes seizures, global developmental delay and intellectual disability. METHODS Two of the patients described herein were identified via genome-wide and exome-wide testing, with microarray and research-based exome, through the CAUSES (Clinical Assessment of the Utility of Sequencing and Evaluation as a Service) Research Clinic at the University of British Columbia. The third Vancouver patient had clinical trio exome sequencing through Blueprint Genetics. The fourth patient underwent singleton exome sequencing in Nantes, with subsequent recruitment to this cohort through GeneMatcher. RESULTS Here we present clinical reports of four patients with rare coding variants in SETD1B that demonstrate a shared phenotype, including intellectual disability, language delay, conserved musculoskeletal findings and seizures that may be treatment-refractory. We include supporting evidence from next-generation sequencing among a cohort of paediatric patients with epilepsy. CONCLUSION Rare coding variants in SETD1B can cause a diagnosable syndrome and could contribute as a risk factor for epilepsy, autism and other neurodevelopmental phenotypes. In the long term, some patients may also be at increased risk for cancers and other complex diseases. Thus, longitudinal studies are required to further elucidate the precise role of SETD1B in neurodevelopmental disorders and other systemic disease.
Collapse
Affiliation(s)
- Alexandra Roston
- Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Dan Evans
- Centre for Applied Neurogenetics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Harinder Gill
- Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada.,Provincial Medical Genetics Program, BC Women's Hospital and Health Centre, Vancouver, British Columbia, Canada
| | - Margaret McKinnon
- Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Bertrand Isidor
- Service de Génétique Médicale, Centre Hospitalier Universitaire de Nantes, Nantes, Pays de la Loire, France
| | - Benjamin Cogné
- Service de Génétique Médicale, Centre Hospitalier Universitaire de Nantes, Nantes, Pays de la Loire, France.,INSERM, CNRS, UNIV Nantes, l'institut du thorax, Nantes, Frances
| | - Jill Mwenifumbo
- Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Clara van Karnebeek
- Department of Pediatrics, Emma Children's Hospital, Amsterdam Gastroenterology and Metabolism, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, Netherlands.,Department of Pediatrics, Radboud Centre for Mitochondrial Medicine, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Jianghong An
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Matthew Farrer
- Centre for Applied Neurogenetics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Michelle Demos
- Division of Neurology, Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mary Connolly
- Division of Neurology, Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - William T Gibson
- Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada
| | | | | |
Collapse
|
154
|
Smeland OB, Frei O, Dale AM, Andreassen OA. The polygenic architecture of schizophrenia — rethinking pathogenesis and nosology. Nat Rev Neurol 2020; 16:366-379. [DOI: 10.1038/s41582-020-0364-0] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2020] [Indexed: 02/07/2023]
|
155
|
Marenne G, Hendricks AE, Perdikari A, Bounds R, Payne F, Keogh JM, Lelliott CJ, Henning E, Pathan S, Ashford S, Bochukova EG, Mistry V, Daly A, Hayward C, Wareham NJ, O'Rahilly S, Langenberg C, Wheeler E, Zeggini E, Farooqi IS, Barroso I. Exome Sequencing Identifies Genes and Gene Sets Contributing to Severe Childhood Obesity, Linking PHIP Variants to Repressed POMC Transcription. Cell Metab 2020; 31:1107-1119.e12. [PMID: 32492392 PMCID: PMC7267775 DOI: 10.1016/j.cmet.2020.05.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/06/2020] [Accepted: 05/09/2020] [Indexed: 12/12/2022]
Abstract
Obesity is genetically heterogeneous with monogenic and complex polygenic forms. Using exome and targeted sequencing in 2,737 severely obese cases and 6,704 controls, we identified three genes (PHIP, DGKI, and ZMYM4) with an excess burden of very rare predicted deleterious variants in cases. In cells, we found that nuclear PHIP (pleckstrin homology domain interacting protein) directly enhances transcription of pro-opiomelanocortin (POMC), a neuropeptide that suppresses appetite. Obesity-associated PHIP variants repressed POMC transcription. Our demonstration that PHIP is involved in human energy homeostasis through transcriptional regulation of central melanocortin signaling has potential diagnostic and therapeutic implications for patients with obesity and developmental delay. Additionally, we found an excess burden of predicted deleterious variants involving genes nearest to loci from obesity genome-wide association studies. Genes and gene sets influencing obesity with variable penetrance provide compelling evidence for a continuum of causality in the genetic architecture of obesity, and explain some of its missing heritability.
Collapse
Affiliation(s)
- Gaëlle Marenne
- Wellcome Sanger Institute, Cambridge, UK; Inserm, Univ Brest, EFS, UMR 1078, GGB, 29200 Brest, France
| | - Audrey E Hendricks
- Wellcome Sanger Institute, Cambridge, UK; Mathematical and Statistical Sciences, University of Colorado Denver, Denver, CO, USA
| | - Aliki Perdikari
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Rebecca Bounds
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | | | - Julia M Keogh
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | | | - Elana Henning
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Saad Pathan
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Sofie Ashford
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Elena G Bochukova
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Vanisha Mistry
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Allan Daly
- Wellcome Sanger Institute, Cambridge, UK
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK; Generation Scotland, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Nicholas J Wareham
- University of Cambridge MRC Epidemiology Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Stephen O'Rahilly
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Claudia Langenberg
- University of Cambridge MRC Epidemiology Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Eleanor Wheeler
- Wellcome Sanger Institute, Cambridge, UK; University of Cambridge MRC Epidemiology Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Eleftheria Zeggini
- Wellcome Sanger Institute, Cambridge, UK; Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - I Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
| | - Inês Barroso
- Wellcome Sanger Institute, Cambridge, UK; University of Cambridge MRC Epidemiology Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
| |
Collapse
|
156
|
Sze CC, Ozark PA, Cao K, Ugarenko M, Das S, Wang L, Marshall SA, Rendleman EJ, Ryan CA, Zha D, Douillet D, Chen FX, Shilatifard A. Coordinated regulation of cellular identity-associated H3K4me3 breadth by the COMPASS family. SCIENCE ADVANCES 2020; 6:eaaz4764. [PMID: 32637595 PMCID: PMC7314515 DOI: 10.1126/sciadv.aaz4764] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 05/08/2020] [Indexed: 06/01/2023]
Abstract
Set1A and Set1B, two members of the COMPASS family of methyltransferases that methylate the histone H3 lysine 4 (H3K4) residue, have been accredited as primary depositors of global H3K4 trimethylation (H3K4me3) in mammalian cells. Our previous studies in mouse embryonic stem cells (ESCs) demonstrated that deleting the enzymatic SET domain of Set1A does not perturb bulk H3K4me3, indicating possible compensatory roles played by other COMPASS methyltransferases. Here, we generated a series of ESC lines harboring compounding mutations of COMPASS methyltransferases. We find that Set1B is functionally redundant to Set1A in implementing H3K4me3 at highly expressed genes, while Mll2 deposits H3K4me3 at less transcriptionally active promoters. While Set1A-B/COMPASS is responsible for broad H3K4me3 peaks, Mll2/COMPASS establishes H3K4me3 with narrow breadth. Additionally, Mll2 helps preserve global H3K4me3 levels and peak breadth in the absence of Set1A-B activity. Our results illustrate the biological flexibility of such enzymes in regulating transcription in a context-dependent manner to maintain stem cell identity.
Collapse
Affiliation(s)
- Christie C. Sze
- Simpson Querrey Center for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, 303 E. Superior St., Chicago, IL 60611, USA
| | - Patrick A. Ozark
- Simpson Querrey Center for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, 303 E. Superior St., Chicago, IL 60611, USA
| | - Kaixiang Cao
- Simpson Querrey Center for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, 303 E. Superior St., Chicago, IL 60611, USA
| | - Michal Ugarenko
- Simpson Querrey Center for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, 303 E. Superior St., Chicago, IL 60611, USA
| | - Siddhartha Das
- Simpson Querrey Center for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, 303 E. Superior St., Chicago, IL 60611, USA
| | - Lu Wang
- Simpson Querrey Center for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, 303 E. Superior St., Chicago, IL 60611, USA
| | - Stacy A. Marshall
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, 303 E. Superior St., Chicago, IL 60611, USA
| | - Emily J. Rendleman
- Simpson Querrey Center for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, 303 E. Superior St., Chicago, IL 60611, USA
| | - Caila A. Ryan
- Simpson Querrey Center for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, 303 E. Superior St., Chicago, IL 60611, USA
| | - Didi Zha
- Simpson Querrey Center for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, 303 E. Superior St., Chicago, IL 60611, USA
| | - Delphine Douillet
- Simpson Querrey Center for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, 303 E. Superior St., Chicago, IL 60611, USA
| | - Fei Xavier Chen
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, 303 E. Superior St., Chicago, IL 60611, USA
| | - Ali Shilatifard
- Simpson Querrey Center for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, 303 E. Superior St., Chicago, IL 60611, USA
- Robert H. Lurie NCI Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Superior St., Chicago, IL 60611, USA
| |
Collapse
|
157
|
Köhler S, Carmody L, Vasilevsky N, Jacobsen JOB, Danis D, Gourdine JP, Gargano M, Harris NL, Matentzoglu N, McMurry JA, Osumi-Sutherland D, Cipriani V, Balhoff JP, Conlin T, Blau H, Baynam G, Palmer R, Gratian D, Dawkins H, Segal M, Jansen AC, Muaz A, Chang WH, Bergerson J, Laulederkind SJF, Yüksel Z, Beltran S, Freeman AF, Sergouniotis PI, Durkin D, Storm AL, Hanauer M, Brudno M, Bello SM, Sincan M, Rageth K, Wheeler MT, Oegema R, Lourghi H, Della Rocca MG, Thompson R, Castellanos F, Priest J, Cunningham-Rundles C, Hegde A, Lovering RC, Hajek C, Olry A, Notarangelo L, Similuk M, Zhang XA, Gómez-Andrés D, Lochmüller H, Dollfus H, Rosenzweig S, Marwaha S, Rath A, Sullivan K, Smith C, Milner JD, Leroux D, Boerkoel CF, Klion A, Carter MC, Groza T, Smedley D, Haendel MA, Mungall C, Robinson PN. Expansion of the Human Phenotype Ontology (HPO) knowledge base and resources. Nucleic Acids Res 2020; 47:D1018-D1027. [PMID: 30476213 PMCID: PMC6324074 DOI: 10.1093/nar/gky1105] [Citation(s) in RCA: 441] [Impact Index Per Article: 88.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 10/24/2018] [Indexed: 12/12/2022] Open
Abstract
The Human Phenotype Ontology (HPO)—a standardized vocabulary of phenotypic abnormalities associated with 7000+ diseases—is used by thousands of researchers, clinicians, informaticians and electronic health record systems around the world. Its detailed descriptions of clinical abnormalities and computable disease definitions have made HPO the de facto standard for deep phenotyping in the field of rare disease. The HPO’s interoperability with other ontologies has enabled it to be used to improve diagnostic accuracy by incorporating model organism data. It also plays a key role in the popular Exomiser tool, which identifies potential disease-causing variants from whole-exome or whole-genome sequencing data. Since the HPO was first introduced in 2008, its users have become both more numerous and more diverse. To meet these emerging needs, the project has added new content, language translations, mappings and computational tooling, as well as integrations with external community data. The HPO continues to collaborate with clinical adopters to improve specific areas of the ontology and extend standardized disease descriptions. The newly redesigned HPO website (www.human-phenotype-ontology.org) simplifies browsing terms and exploring clinical features, diseases, and human genes.
Collapse
Affiliation(s)
- Sebastian Köhler
- Charité Centrum für Therapieforschung, Charité-Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin 10117, Germany.,Einstein Center Digital Future, Berlin 10117, Germany.,Monarch Initiative, monarchinitiative.org
| | - Leigh Carmody
- Monarch Initiative, monarchinitiative.org.,The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Nicole Vasilevsky
- Monarch Initiative, monarchinitiative.org.,Oregon Health & Science University, Portland, OR 97217, USA
| | - Julius O B Jacobsen
- Monarch Initiative, monarchinitiative.org.,Genomics England, Queen Mary University of London, Dawson Hall, Charterhouse Square, London EC1M 6BQ, UK
| | - Daniel Danis
- Monarch Initiative, monarchinitiative.org.,The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Jean-Philippe Gourdine
- Monarch Initiative, monarchinitiative.org.,Oregon Health & Science University, Portland, OR 97217, USA
| | - Michael Gargano
- Monarch Initiative, monarchinitiative.org.,The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Nomi L Harris
- Monarch Initiative, monarchinitiative.org.,Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Nicolas Matentzoglu
- Monarch Initiative, monarchinitiative.org.,European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Cambridge, UK
| | - Julie A McMurry
- Monarch Initiative, monarchinitiative.org.,Linus Pauling institute, Oregon State University, Corvallis, OR, USA
| | - David Osumi-Sutherland
- Monarch Initiative, monarchinitiative.org.,European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Cambridge, UK
| | - Valentina Cipriani
- Monarch Initiative, monarchinitiative.org.,William Harvey Research Institute, Queen Mary University College of London.,UCL Genetics Institute, University College of London.,UCL Institute of Ophthalmology, University College of London
| | - James P Balhoff
- Monarch Initiative, monarchinitiative.org.,Renaissance Computing Institute, University of North Carolina at Chapel Hill
| | - Tom Conlin
- Monarch Initiative, monarchinitiative.org.,Linus Pauling institute, Oregon State University, Corvallis, OR, USA
| | - Hannah Blau
- Monarch Initiative, monarchinitiative.org.,The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Gareth Baynam
- Western Australian Register of Developmental Anomalies and Genetic Services of Western Australia, Department of Health, Government of Western Australia, WA, Australia.,School of Paediatrics and Telethon Kids Institute, University of Western Australia, Perth, WA, Australia.,Institute for Immunology and Infectious Diseases, Murdoch University, Perth, WA, Australia.,Spatial Sciences, Department of Science and Engineering, Curtin University, Perth, WA, Australia.,The Office of Population Health Genomics, Department of Health, Government of Western Australia, Perth, WA, Australia
| | - Richard Palmer
- Spatial Sciences, Department of Science and Engineering, Curtin University, Perth, WA, Australia
| | - Dylan Gratian
- Western Australian Register of Developmental Anomalies and Genetic Services of Western Australia, Department of Health, Government of Western Australia, WA, Australia
| | - Hugh Dawkins
- The Office of Population Health Genomics, Department of Health, Government of Western Australia, Perth, WA, Australia
| | | | - Anna C Jansen
- Neurogenetics Research Group, Vrije Universiteit Brussel, Brussels, Belgium.,Pediatric Neurology Unit, Department of Pediatrics, UZ Brussel, Brussels, Belgium
| | - Ahmed Muaz
- Monarch Initiative, monarchinitiative.org.,Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW 2010, Australia
| | - Willie H Chang
- Centre for Computational Medicine, Hospital for Sick Children and Department of Computer Science, University of Toronto, Toronto, Canada
| | - Jenna Bergerson
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stanley J F Laulederkind
- Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin & Marquette University, 8701 Watertown Plank Road Milwaukee, WI 53226, USA
| | | | - Sergi Beltran
- CNAG-CRG, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Baldiri Reixac 4, Barcelona 08028, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Alexandra F Freeman
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Daniel Durkin
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Andrea L Storm
- ICF, Rockville, MD, USA.,National Center for Advancing Translational Sciences, Office of Rare Diseases Research, National Institutes of Health, Bethesda, MD, USA
| | - Marc Hanauer
- INSERM, US14-Orphanet, Plateforme Maladies Rares, 75014 Paris, France
| | - Michael Brudno
- Centre for Computational Medicine, Hospital for Sick Children and Department of Computer Science, University of Toronto, Toronto, Canada
| | | | - Murat Sincan
- Sanford Imagenetics, Sanford Health, Sioux Falls, SD, USA
| | - Kayli Rageth
- Sanford Imagenetics, Sanford Health, Sioux Falls, SD, USA
| | - Matthew T Wheeler
- Center for Undiagnosed Diseases, Stanford University School of Medicine, Stanford, CA, USA
| | - Renske Oegema
- Department of Genetics, University Medical Center Utrecht, the Netherlands
| | - Halima Lourghi
- INSERM, US14-Orphanet, Plateforme Maladies Rares, 75014 Paris, France
| | - Maria G Della Rocca
- ICF, Rockville, MD, USA.,National Center for Advancing Translational Sciences, Office of Rare Diseases Research, National Institutes of Health, Bethesda, MD, USA
| | - Rachel Thompson
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | | | - James Priest
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Ayushi Hegde
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Ruth C Lovering
- Institute of Cardiovascular Science, University College London, UK
| | | | - Annie Olry
- INSERM, US14-Orphanet, Plateforme Maladies Rares, 75014 Paris, France
| | - Luigi Notarangelo
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Morgan Similuk
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Xingmin A Zhang
- Monarch Initiative, monarchinitiative.org.,The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - David Gómez-Andrés
- Child Neurology Unit. Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Hanns Lochmüller
- CNAG-CRG, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Baldiri Reixac 4, Barcelona 08028, Spain.,Department of Neuropediatrics and Muscle Disorders, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany.,Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Canada.,Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, Canada
| | - Hélène Dollfus
- Centre for Rare Eye Diseases CARGO, SENSGENE FSMR Network, Strasbourg University Hospital, Strasbourg, France
| | - Sergio Rosenzweig
- Immunology Service, Department of Laboratory Medicine, NIH Clinical Center, Bethesda, MD, USA
| | - Shruti Marwaha
- Center for Undiagnosed Diseases, Stanford University School of Medicine, Stanford, CA, USA
| | - Ana Rath
- INSERM, US14-Orphanet, Plateforme Maladies Rares, 75014 Paris, France
| | - Kathleen Sullivan
- Department of Pediatrics, Division of Allergy Immunology, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, 3615 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | | | - Joshua D Milner
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Dorothée Leroux
- Centre for Rare Eye Diseases CARGO, SENSGENE FSMR Network, Strasbourg University Hospital, Strasbourg, France
| | | | - Amy Klion
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Melody C Carter
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tudor Groza
- Monarch Initiative, monarchinitiative.org.,Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW 2010, Australia
| | - Damian Smedley
- Monarch Initiative, monarchinitiative.org.,Genomics England, Queen Mary University of London, Dawson Hall, Charterhouse Square, London EC1M 6BQ, UK
| | - Melissa A Haendel
- Monarch Initiative, monarchinitiative.org.,Oregon Health & Science University, Portland, OR 97217, USA.,Linus Pauling institute, Oregon State University, Corvallis, OR, USA
| | - Chris Mungall
- Monarch Initiative, monarchinitiative.org.,Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Peter N Robinson
- Monarch Initiative, monarchinitiative.org.,The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA.,Institute for Systems Genomics, University of Connecticut, Farmington, CT, USA
| |
Collapse
|
158
|
Genetic and epigenetic analyses of panic disorder in the post-GWAS era. J Neural Transm (Vienna) 2020; 127:1517-1526. [PMID: 32388794 PMCID: PMC7578165 DOI: 10.1007/s00702-020-02205-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/03/2020] [Indexed: 02/07/2023]
Abstract
Panic disorder (PD) is a common and debilitating neuropsychiatric disorder characterized by panic attacks coupled with excessive anxiety. Both genetic factors and environmental factors play an important role in PD pathogenesis and response to treatment. However, PD is clinically heterogeneous and genetically complex, and the exact genetic or environmental causes of this disorder remain unclear. Various approaches for detecting disease-causing genes have recently been made available. In particular, genome-wide association studies (GWAS) have attracted attention for the identification of disease-associated loci of multifactorial disorders. This review introduces GWAS of PD, followed by a discussion about the limitations of GWAS and the major challenges facing geneticists in the post-GWAS era. Alternative strategies to address these challenges are then proposed, such as epigenome-wide association studies (EWAS) and rare variant association studies (RVAS) using next-generation sequencing. To date, however, few reports have described these analyses, and the evidence remains insufficient to confidently identify or exclude rare variants or epigenetic changes in PD. Further analyses are therefore required, using sample sizes in the tens of thousands, extensive functional annotations, and highly targeted hypothesis testing.
Collapse
|
159
|
Kranz A, Anastassiadis K. The role of SETD1A and SETD1B in development and disease. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2020; 1863:194578. [PMID: 32389824 DOI: 10.1016/j.bbagrm.2020.194578] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/16/2020] [Accepted: 05/03/2020] [Indexed: 12/13/2022]
Abstract
The Trithorax-related Set1 H3K4 methyltransferases are conserved from yeast to human. In yeast loss of Set1 causes pleiotropic effects but is compatible with life. In contrast, both mammalian Set1 orthologs: SETD1A and SETD1B are essential for embryonic development, however they have distinct functions. SETD1A is required shortly after epiblast formation whereas SETD1B becomes indispensible during early organogenesis. In adult mice both SETD1A and SETD1B regulate hematopoiesis differently: SETD1A is required for the establishment of definitive hematopoiesis whereas SETD1B is important for the maintenance of long-term hematopoietic stem cells. Both are implicated in different diseases with accumulating evidence for the association of SETD1A variants in neurological disorders and SETD1B variants with cancer. Why the two paralogs cannot or only partially compensate for the loss of each other is part of the puzzle that we try to sort out in this review.
Collapse
Affiliation(s)
- Andrea Kranz
- Genomics, Center for Molecular and Cellular Bioengineering, Biotechnology Center, Technische Universität Dresden, Tatzberg 47, 01307 Dresden, Germany
| | - Konstantinos Anastassiadis
- Stem Cell Engineering, Center for Molecular and Cellular Bioengineering, Biotechnology Center, Technische Universität Dresden, Tatzberg 47, 01307 Dresden, Germany.
| |
Collapse
|
160
|
Levchenko A, Nurgaliev T, Kanapin A, Samsonova A, Gainetdinov RR. Current challenges and possible future developments in personalized psychiatry with an emphasis on psychotic disorders. Heliyon 2020; 6:e03990. [PMID: 32462093 PMCID: PMC7240336 DOI: 10.1016/j.heliyon.2020.e03990] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 10/31/2019] [Accepted: 05/12/2020] [Indexed: 12/13/2022] Open
Abstract
A personalized medicine approach seems to be particularly applicable to psychiatry. Indeed, considering mental illness as deregulation, unique to each patient, of molecular pathways, governing the development and functioning of the brain, seems to be the most justified way to understand and treat disorders of this medical category. In order to extract correct information about the implicated molecular pathways, data can be drawn from sampling phenotypic and genetic biomarkers and then analyzed by a machine learning algorithm. This review describes current difficulties in the field of personalized psychiatry and gives several examples of possibly actionable biomarkers of psychotic and other psychiatric disorders, including several examples of genetic studies relevant to personalized psychiatry. Most of these biomarkers are not yet ready to be introduced in clinical practice. In a next step, a perspective on the path personalized psychiatry may take in the future is given, paying particular attention to machine learning algorithms that can be used with the goal of handling multidimensional datasets.
Collapse
Affiliation(s)
- Anastasia Levchenko
- Theodosius Dobzhansky Center for Genome Bioinformatics, Saint Petersburg State University, 7/9 Universitetskaya nab., Saint Petersburg, 199034, Russia
| | - Timur Nurgaliev
- Institute of Translational Biomedicine, Saint Petersburg State University, 7/9 Universitetskaya nab., Saint Petersburg, 199034, Russia
| | - Alexander Kanapin
- Theodosius Dobzhansky Center for Genome Bioinformatics, Saint Petersburg State University, 7/9 Universitetskaya nab., Saint Petersburg, 199034, Russia
| | - Anastasia Samsonova
- Theodosius Dobzhansky Center for Genome Bioinformatics, Saint Petersburg State University, 7/9 Universitetskaya nab., Saint Petersburg, 199034, Russia
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, Saint Petersburg State University, 7/9 Universitetskaya nab., Saint Petersburg, 199034, Russia
| |
Collapse
|
161
|
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
|
162
|
Zhu Y, Sun D, Jakovcevski M, Jiang Y. Epigenetic mechanism of SETDB1 in brain: implications for neuropsychiatric disorders. Transl Psychiatry 2020; 10:115. [PMID: 32321908 PMCID: PMC7176658 DOI: 10.1038/s41398-020-0797-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 03/13/2020] [Accepted: 03/26/2020] [Indexed: 12/14/2022] Open
Abstract
Neuropsychiatric disorders are a collective of cerebral conditions with a multifactorial and polygenetic etiology. Dysregulation of epigenetic profiles in the brain is considered to play a critical role in the development of neuropsychiatric disorders. SET domain, bifurcate 1 (SETDB1), functioning as a histone H3K9 specific methyltransferase, is not only critically involved in transcriptional silencing and local heterochromatin formation, but also affects genome-wide neuronal epigenetic profiles and is essential for 3D genome integrity. Here, we provide a review of recent advances towards understanding the role of SETDB1 in the central nervous system during early neurodevelopment as well as in the adult brain, with a particular focus on studies that link its functions to neuropsychiatric disorders and related behavioral changes, and the exploration of novel therapeutic strategies targeting SETDB1.
Collapse
Affiliation(s)
- Yueyan Zhu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Fudan University, 200032, Shanghai, China
| | - Daijing Sun
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Fudan University, 200032, Shanghai, China
| | - Mira Jakovcevski
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Yan Jiang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Fudan University, 200032, Shanghai, China.
| |
Collapse
|
163
|
Seah C, Brennand KJ. If there is not one cure for schizophrenia, there may be many. NPJ SCHIZOPHRENIA 2020; 6:11. [PMID: 32313122 PMCID: PMC7170875 DOI: 10.1038/s41537-020-0101-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/28/2020] [Indexed: 11/24/2022]
Affiliation(s)
- Carina Seah
- Graduate School of Biomedical Science, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kristen J Brennand
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
164
|
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
|
165
|
Jiang H. The complex activities of the SET1/MLL complex core subunits in development and disease. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2020; 1863:194560. [PMID: 32302696 DOI: 10.1016/j.bbagrm.2020.194560] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/14/2020] [Accepted: 04/09/2020] [Indexed: 12/14/2022]
Abstract
In mammalian cells, the SET1/MLL complexes are the main writers of the H3K4 methyl mark that is associated with active gene expression. The activities of these complexes are critically dependent on the association of the catalytic subunit with their shared core subunits, WDR5, RBBP5, ASH2L, and DPY30, collectively referred as WRAD. In addition, some of these core subunits can bind to proteins other than the SET1/MLL complex components. This review starts with discussion of the molecular activities of these core subunits, with an emphasis on DPY30 in organizing the assembly of the SET1/MLL complexes with other associated factors. This review then focuses on the roles of the core subunits in stem cells and development, as well as in diseased cell states, mainly cancer, and ends with discussion on dissecting the responsible activities of the core subunits and how we may target them for potential disease treatment. This article is part of a Special Issue entitled: The MLL family of proteins in normal development and disease edited by Thomas A Milne.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| |
Collapse
|
166
|
Sey NYA, Hu B, Mah W, Fauni H, McAfee JC, Rajarajan P, Brennand KJ, Akbarian S, Won H. A computational tool (H-MAGMA) for improved prediction of brain-disorder risk genes by incorporating brain chromatin interaction profiles. Nat Neurosci 2020; 23:583-593. [PMID: 32152537 PMCID: PMC7131892 DOI: 10.1038/s41593-020-0603-0] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 01/30/2020] [Indexed: 11/23/2022]
Abstract
Most risk variants for brain disorders identified by genome-wide association studies (GWAS) reside in non-coding genome, which makes deciphering biological mechanisms difficult. A commonly used tool, MAGMA, addresses this issue by aggregating SNP associations to nearest genes. Here, we developed a platform, Hi-C coupled MAGMA (H-MAGMA), that advances MAGMA by incorporating chromatin interaction profiles from human brain tissue across two developmental epochs and two brain cell types. By employing gene regulatory relationships in the disease-relevant tissue, H-MAGMA identifies neurobiologically-relevant target genes. We applied H-MAGMA to five psychiatric disorders and four neurodegenerative disorders to interrogate biological pathways, developmental windows, and cell types implicated for each disorder. Psychiatric disorder risk genes tended to be expressed during mid-gestation and in excitatory neurons, whereas degenerative disorder risk genes showed increasing expression over time and more diverse cell-type specificities. H-MAGMA adds to existing analytic frameworks to help identify the neurobiological consequences of brain disorder genetics.
Collapse
Affiliation(s)
- Nancy Y A Sey
- UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA
| | - Benxia Hu
- UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA.,Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Won Mah
- UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA.,Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Harper Fauni
- UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA
| | - Jessica Caitlin McAfee
- UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA.,Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Prashanth Rajarajan
- Graduate School of Biomedical Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristen J Brennand
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Schahram Akbarian
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hyejung Won
- UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA. .,Department of Genetics, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
167
|
Koch N, Koch D, Krueger S, Tröger J, Sabanov V, Ahmed T, McMillan LE, Wolf D, Montag D, Kessels MM, Balschun D, Qualmann B. Syndapin I Loss-of-Function in Mice Leads to Schizophrenia-Like Symptoms. Cereb Cortex 2020; 30:4306-4324. [DOI: 10.1093/cercor/bhaa013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 12/12/2022] Open
Abstract
Abstract
Schizophrenia is associated with cognitive and behavioral dysfunctions thought to reflect imbalances in neurotransmission systems. Recent screenings suggested that lack of (functional) syndapin I (PACSIN1) may be linked to schizophrenia. We therefore studied syndapin I KO mice to address the suggested causal relationship to schizophrenia and to analyze associated molecular, cellular, and neurophysiological defects. Syndapin I knockout (KO) mice developed schizophrenia-related behaviors, such as hyperactivity, reduced anxiety, reduced response to social novelty, and an exaggerated novel object response and exhibited defects in dendritic arborization in the cortex. Neuromorphogenic deficits were also observed for a schizophrenia-associated syndapin I mutant in cultured neurons and coincided with a lack of syndapin I–mediated membrane recruitment of cytoskeletal effectors. Syndapin I KO furthermore caused glutamatergic hypofunctions. Syndapin I regulated both AMPAR and NMDAR availabilities at synapses during basal synaptic activity and during synaptic plasticity—particularly striking were a complete lack of long-term potentiation and defects in long-term depression in syndapin I KO mice. These synaptic plasticity defects coincided with alterations of postsynaptic actin dynamics, synaptic GluA1 clustering, and GluA1 mobility. Both GluA1 and GluA2 were not appropriately internalized. Summarized, syndapin I KO led to schizophrenia-like behavior, and our analyses uncovered associated molecular and cellular mechanisms.
Collapse
Affiliation(s)
- Nicole Koch
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Dennis Koch
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Sarah Krueger
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Jessica Tröger
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Victor Sabanov
- Laboratory of Biological Psychology, Brain & Cognition, University of Leuven, 3000 Leuven, Belgium
| | - Tariq Ahmed
- Laboratory of Biological Psychology, Brain & Cognition, University of Leuven, 3000 Leuven, Belgium
| | - Laura E McMillan
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, 07743 Jena, Germany
| | - David Wolf
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Dirk Montag
- Neurogenetics Lab, Leibniz Institute for Neurobiology, 39116 Magdeburg, Germany
| | - Michael M Kessels
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Detlef Balschun
- Laboratory of Biological Psychology, Brain & Cognition, University of Leuven, 3000 Leuven, Belgium
| | - Britta Qualmann
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, 07743 Jena, Germany
| |
Collapse
|
168
|
Gogos JA, Crabtree G, Diamantopoulou A. The abiding relevance of mouse models of rare mutations to psychiatric neuroscience and therapeutics. Schizophr Res 2020; 217:37-51. [PMID: 30987923 PMCID: PMC6790166 DOI: 10.1016/j.schres.2019.03.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/19/2019] [Accepted: 03/22/2019] [Indexed: 01/08/2023]
Abstract
Studies using powerful family-based designs aided by large scale case-control studies, have been instrumental in cracking the genetic complexity of the disease, identifying rare and highly penetrant risk mutations and providing a handle on experimentally tractable model systems. Mouse models of rare mutations, paired with analysis of homologous cognitive and sensory processing deficits and state-of-the-art neuroscience methods to manipulate and record neuronal activity have started providing unprecedented insights into pathogenic mechanisms and building the foundation of a new biological framework for understanding mental illness. A number of important principles are emerging, namely that degradation of the computational mechanisms underlying the ordered activity and plasticity of both local and long-range neuronal assemblies, the building blocks necessary for stable cognition and perception, might be the inevitable consequence and the common point of convergence of the vastly heterogeneous genetic liability, manifesting as defective internally- or stimulus-driven neuronal activation patterns and triggering the constellation of schizophrenia symptoms. Animal models of rare mutations have the unique potential to help us move from "which" (gene) to "how", "where" and "when" computational regimes of neural ensembles are affected. Linking these variables should improve our understanding of how symptoms emerge and how diagnostic boundaries are established at a circuit level. Eventually, a better understanding of pathophysiological trajectories at the level of neural circuitry in mice, aided by basic human experimental biology, should guide the development of new therapeutics targeting either altered circuitry itself or the underlying biological pathways.
Collapse
Affiliation(s)
- Joseph A. Gogos
- Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027 USA,Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA,Department of Neuroscience, Columbia University, New York, NY 10032 USA,Correspondence should be addressed to: Joseph A. Gogos ()
| | - Gregg Crabtree
- Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027 USA,Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Anastasia Diamantopoulou
- Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027 USA,Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| |
Collapse
|
169
|
Rajarajan P, Flaherty E, Akbarian S, Brennand KJ. CRISPR-based functional evaluation of schizophrenia risk variants. Schizophr Res 2020; 217:26-36. [PMID: 31277978 PMCID: PMC6939156 DOI: 10.1016/j.schres.2019.06.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [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: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 02/06/2023]
Abstract
As expanding genetic and genomic studies continue to implicate a growing list of variants contributing risk to neuropsychiatric disease, an important next step is to understand the functional impact and points of convergence of these risk factors. Here, with a focus on schizophrenia, we survey the most recent findings of the rare and common variants underlying genetic risk for schizophrenia. We discuss the ongoing efforts to validate these variants in post-mortem brain tissue, as well as new approaches to combine CRISPR-based genome engineering with patient-specific human induced pluripotent stem cell (hiPSC)-based models, in order to identify putative causal schizophrenia loci that regulate gene expression and cellular function. We consider the current limitations of hiPSC-based approaches as well as the future advances necessary to improve the fidelity of this human model. With the objective of utilizing patient genotype data to improve diagnosis and predict treatment response, the integration of CRISPR-genome engineering and hiPSC-based models represent an important strategy with which to systematically demonstrate the cell-type-specific effects of schizophrenia-associated variants.
Collapse
Affiliation(s)
- Prashanth Rajarajan
- Graduate School of Biomedical Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America
| | - Erin Flaherty
- Graduate School of Biomedical Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America
| | - Schahram Akbarian
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America
| | - Kristen J Brennand
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America.
| |
Collapse
|
170
|
Mah W, Won H. The three-dimensional landscape of the genome in human brain tissue unveils regulatory mechanisms leading to schizophrenia risk. Schizophr Res 2020; 217:17-25. [PMID: 30894290 PMCID: PMC6748876 DOI: 10.1016/j.schres.2019.03.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 02/27/2019] [Accepted: 03/06/2019] [Indexed: 12/18/2022]
Abstract
Recent advances in our understanding of the genetic architecture of schizophrenia have shed light on the schizophrenia etiology. While common variation is one of the major genetic contributors, the majority of common variation reside in non-coding genome, posing a significant challenge in understanding the functional impact of this class of genetic variation. Functional genomic datasets that range from expression quantitative trait loci (eQTL) to chromatin interactions are critical to identify the potential target genes and functional consequences of non-coding variation. In this review, we discuss how three-dimensional chromatin landscape, identified by a technique called Hi-C, has facilitated the identification of potential target genes impacting schizophrenia risk. We outline key steps for Hi-C driven gene mapping, and compare Hi-C defined schizophrenia risk genes defined across developmental epochs and cell types, which offer rich insights into the temporal window and cellular etiology of schizophrenia. In contrast with a neurodevelopmental hypothesis in schizophrenia, Hi-C defined schizophrenia risk genes are postnatally enriched, suggesting that postnatal development is also important for schizophrenia pathogenesis. Moreover, Hi-C defined schizophrenia risk genes are highly expressed in excitatory neurons, highlighting excitatory neurons as a central cell type for schizophrenia. Further characterization of Hi-C defined schizophrenia risk genes demonstrated enrichment for genes that harbor loss-of-function variation in neurodevelopmental disorders, suggesting a shared genetic etiology between schizophrenia and neurodevelopmental disorders. Collectively, moving the search space from risk variants to the target genes lays a foundation to understand the neurobiological basis of schizophrenia.
Collapse
Affiliation(s)
- Won Mah
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Hyejung Won
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
171
|
Riveros-Mckay F, Oliver-Williams C, Karthikeyan S, Walter K, Kundu K, Ouwehand WH, Roberts D, Di Angelantonio E, Soranzo N, Danesh J, INTERVAL Study, Wheeler E, Zeggini E, Butterworth AS, Barroso I. The influence of rare variants in circulating metabolic biomarkers. PLoS Genet 2020; 16:e1008605. [PMID: 32150548 PMCID: PMC7108731 DOI: 10.1371/journal.pgen.1008605] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 03/31/2020] [Accepted: 01/10/2020] [Indexed: 12/19/2022] Open
Abstract
Circulating metabolite levels are biomarkers for cardiovascular disease (CVD). Here we studied, association of rare variants and 226 serum lipoproteins, lipids and amino acids in 7,142 (discovery plus follow-up) healthy participants. We leveraged the information from multiple metabolite measurements on the same participants to improve discovery in rare variant association analyses for gene-based and gene-set tests by incorporating correlated metabolites as covariates in the validation stage. Gene-based analysis corrected for the effective number of tests performed, confirmed established associations at APOB, APOC3, PAH, HAL and PCSK (p<1.32x10-7) and identified novel gene-trait associations at a lower stringency threshold with ACSL1, MYCN, FBXO36 and B4GALNT3 (p<2.5x10-6). Regulation of the pyruvate dehydrogenase (PDH) complex was associated for the first time, in gene-set analyses also corrected for effective number of tests, with IDL and LDL parameters, as well as circulating cholesterol (pMETASKAT<2.41x10-6). In conclusion, using an approach that leverages metabolite measurements obtained in the same participants, we identified novel loci and pathways involved in the regulation of these important metabolic biomarkers. As large-scale biobanks continue to amass sequencing and phenotypic information, analytical approaches such as ours will be useful to fully exploit the copious amounts of biological data generated in these efforts.
Collapse
Affiliation(s)
| | - Clare Oliver-Williams
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- Homerton College, Cambridge, United Kingdom
| | - Savita Karthikeyan
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | | | - Kousik Kundu
- Wellcome Sanger Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Willem H. Ouwehand
- Wellcome Sanger Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - David Roberts
- The National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- NHS Blood and Transplant—Oxford Centre, Level 2, John Radcliffe Hospital, Oxford, United Kingdom
- Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Emanuele Di Angelantonio
- Wellcome Sanger Institute, Cambridge, United Kingdom
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- The National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, United Kingdom
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, United Kingdom
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, United Kingdom
| | - Nicole Soranzo
- Wellcome Sanger Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - John Danesh
- Wellcome Sanger Institute, Cambridge, United Kingdom
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- The National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, United Kingdom
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, United Kingdom
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, United Kingdom
| | | | - Eleanor Wheeler
- Wellcome Sanger Institute, Cambridge, United Kingdom
- MRC Epidemiology Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Eleftheria Zeggini
- Wellcome Sanger Institute, Cambridge, United Kingdom
- Institute of Translational Genomics, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
| | - Adam S. Butterworth
- Wellcome Sanger Institute, Cambridge, United Kingdom
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- The National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, United Kingdom
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, United Kingdom
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, United Kingdom
| | - Inês Barroso
- Wellcome Sanger Institute, Cambridge, United Kingdom
- MRC Epidemiology Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, United Kingdom
| |
Collapse
|
172
|
Hoya S, Watanabe Y, Nunokawa A, Otsuka I, Shibuya M, Igeta H, Hishimoto A, Someya T. Whole-exome sequencing in a family with a monozygotic twin pair concordant for schizophrenia and a follow-up case-control study of identified de-novo variants. Psychiatr Genet 2020; 30:60-63. [PMID: 32106127 DOI: 10.1097/ypg.0000000000000250] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Whole-exome sequencing (WES) studies have shown that de-novo variants contribute to the genetic etiology of schizophrenia. WES studies of families with a monozygotic twin pair concordant or discordant for a disease may be fruitful for identifying de-novo pathogenic variants. Here, we performed WES in six individuals from one family (affected monozygotic twins, their unaffected parents, and two siblings) and identified three de-novo missense variants (CPT2 Ala283Thr, CPSF3 Val584Ile, and RNF148 Val210Ile) in the monozygotic twin pair concordant for schizophrenia. These three missense variants were not found in 1760 patients with schizophrenia or schizoaffective disorder or 1508 healthy controls. Our data do not support the role of the three missense variants in conferring risk for schizophrenia.
Collapse
Affiliation(s)
- Satoshi Hoya
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences
| | - Yuichiro Watanabe
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences
| | - Ayako Nunokawa
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences.,Minamihama Hospital, Niigata
| | - Ikuo Otsuka
- Department of Psychiatry, Kobe University Graduate School of Medicine, Kobe, Hyogo
| | - Masako Shibuya
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences.,Minamihama Hospital, Niigata
| | - Hirofumi Igeta
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences.,Mano Mizuho Hospital, Sado, Niigata
| | - Akitoyo Hishimoto
- Department of Psychiatry, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Toshiyuki Someya
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences
| |
Collapse
|
173
|
Abstract
BACKGROUND There is increasing evidence that certain genetic variants increase the risk of schizophrenia and other neurodevelopmental disorders. Exome sequencing has been shown to have a high diagnostic yield for developmental disability and testing for copy number variants has been advocated for schizophrenia. The diagnostic yield for exome sequencing in schizophrenia is unknown. METHOD A sample of 591 exome-sequenced schizophrenia cases and their parents were screened for disruptive and damaging variants in autosomal genes listed in the Genomics England panels for intellectual disability and other neurological disorders. RESULTS Previously reported disruptive de novo variants were noted in SETD1A, POGZ, SCN2A, and ZMYND11. Although the loss of function of ZMYND11 is a recognized cause of intellectual disability, it has not previously been noted as a risk factor for schizophrenia. A damaging de novo variant of uncertain significance was noted in NRXN1. A previously reported homozygous damaging variant in BLM is predicted to cause Bloom syndrome in 1 case and 1 case was homozygous for a damaging variant in MCPH1, a result of uncertain significance. There were more than 400 disruptive and damaging variants in the target genes in cases but similar numbers were seen among untransmitted parental alleles and none appeared to be clinically significant. CONCLUSIONS The diagnostic yield from exome sequencing in schizophrenia is low. Disruptive and damaging variants seen in known neuropsychiatric genes should not be automatically assumed to have an etiological role if observed in a patient with schizophrenia.
Collapse
Affiliation(s)
| | - David Curtis
- UCL Genetics Institute, University College London, UK
- Centre for Psychiatry, Barts and the London School of Medicine and Dentistry, UK
| |
Collapse
|
174
|
Rees E, Han J, Morgan J, Carrera N, Escott-Price V, Pocklington AJ, Duffield M, Hall LS, Legge SE, Pardiñas AF, Richards AL, Roth J, Lezheiko T, Kondratyev N, Kaleda V, Golimbet V, Parellada M, González-Peñas J, Arango C, Gawlik M, Kirov G, Walters JTR, Holmans P, O'Donovan MC, Owen MJ. De novo mutations identified by exome sequencing implicate rare missense variants in SLC6A1 in schizophrenia. Nat Neurosci 2020; 23:179-184. [PMID: 31932766 PMCID: PMC7007300 DOI: 10.1038/s41593-019-0565-2] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 11/22/2019] [Indexed: 12/30/2022]
Abstract
Schizophrenia is a highly polygenic disorder with important contributions from both common and rare risk alleles. We analyzed exome sequencing data for de novo variants (DNVs) in a new sample of 613 schizophrenia trios and combined this with published data to give a total of 3,444 trios. In this new data, loss-of-function (LoF) DNVs were significantly enriched among 3,471 LoF-intolerant genes, which supports previous findings. In the full dataset, genes associated with neurodevelopmental disorders (n = 159) were significantly enriched for LoF DNVs. Within these neurodevelopmental disorder genes, SLC6A1, which encodes a γ-aminobutyric acid transporter, was associated with missense-damaging DNVs. In 1,122 trios for which genome-wide common variant data were available, schizophrenia and bipolar disorder polygenic risk were significantly overtransmitted to probands. Probands carrying LoF or deletion DNVs in LoF-intolerant or neurodevelopmental disorder genes had significantly less overtransmission of schizophrenia polygenic risk than did non-carriers, which provides a second robust line of evidence that these DNVs increase liability to schizophrenia.
Collapse
Affiliation(s)
- Elliott Rees
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Jun Han
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Joanne Morgan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Noa Carrera
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Valentina Escott-Price
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Andrew J Pocklington
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Madeleine Duffield
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Lynsey S Hall
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Sophie E Legge
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Antonio F Pardiñas
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Alexander L Richards
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Julian Roth
- Department of Psychiatry and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - Tatyana Lezheiko
- Clinical Genetics Laboratory, Mental Health Research Centre, Moscow, Russia
| | - Nikolay Kondratyev
- Clinical Genetics Laboratory, Mental Health Research Centre, Moscow, Russia
| | - Vasilii Kaleda
- Clinical Genetics Laboratory, Mental Health Research Centre, Moscow, Russia
| | - Vera Golimbet
- Clinical Genetics Laboratory, Mental Health Research Centre, Moscow, Russia
| | - Mara Parellada
- Department of Child and Adolescent Psychiatry, Hospital General Universitario Gregorio Marañón, IisGM, School of Medicine, Universidad Complutense, CIBERSAM, Madrid, Spain
| | - Javier González-Peñas
- Department of Child and Adolescent Psychiatry, Hospital General Universitario Gregorio Marañón, IisGM, School of Medicine, Universidad Complutense, CIBERSAM, Madrid, Spain
| | - Celso Arango
- Department of Child and Adolescent Psychiatry, Hospital General Universitario Gregorio Marañón, IisGM, School of Medicine, Universidad Complutense, CIBERSAM, Madrid, Spain
| | - Micha Gawlik
- Department of Psychiatry and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - George Kirov
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - James T R Walters
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Peter Holmans
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Michael C O'Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK.
| | - Michael J Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK.
| |
Collapse
|
175
|
Howrigan DP, Rose SA, Samocha KE, Fromer M, Cerrato F, Chen WJ, Churchhouse C, Chambert K, Chandler SD, Daly MJ, Dumont A, Genovese G, Hwu HG, Laird N, Kosmicki JA, Moran JL, Roe C, Singh T, Wang SH, Faraone SV, Glatt SJ, McCarroll SA, Tsuang M, Neale BM. Exome sequencing in schizophrenia-affected parent-offspring trios reveals risk conferred by protein-coding de novo mutations. Nat Neurosci 2020; 23:185-193. [PMID: 31932770 PMCID: PMC7007385 DOI: 10.1038/s41593-019-0564-3] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 11/22/2019] [Indexed: 12/31/2022]
Abstract
Protein-coding de novo mutations (DNMs) are significant risk factors in many neurodevelopmental disorders, whereas schizophrenia (SCZ) risk associated with DNMs has thus far been shown to be modest. We analyzed DNMs from 1,695 SCZ-affected trios and 1,077 published SCZ-affected trios to better understand the contribution to SCZ risk. Among 2,772 SCZ probands, exome-wide DNM burden remained modest. Gene set analyses revealed that SCZ DNMs were significantly concentrated in genes that were highly expressed in the brain, that were under strong evolutionary constraint and/or overlapped with genes identified in other neurodevelopmental disorders. No single gene surpassed exome-wide significance; however, 16 genes were recurrently hit by protein-truncating DNMs, corresponding to a 3.15-fold higher rate than the mutation model expectation (permuted 95% confidence interval: 1-10 genes; permuted P = 3 × 10-5). Overall, DNMs explain a small fraction of SCZ risk, and larger samples are needed to identify individual risk genes, as coding variation across many genes confers risk for SCZ in the population.
Collapse
Affiliation(s)
- Daniel P Howrigan
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Samuel A Rose
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kaitlin E Samocha
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Menachem Fromer
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Wei J Chen
- National Taiwan University, Taipei, Taiwan
| | - Claire Churchhouse
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Mark J Daly
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ashley Dumont
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Nan Laird
- Harvard School of Public Health, Boston, MA, USA
| | - Jack A Kosmicki
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Cheryl Roe
- SUNY Upstate Medical University, Syracuse, NY, USA
| | - Tarjinder Singh
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | | | - Steven A McCarroll
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard University, Cambridge, MA, USA
| | - Ming Tsuang
- University of California, San Diego, La Jolla, CA, USA
| | - Benjamin M Neale
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
176
|
Gulsuner S, Stein DJ, Susser ES, Sibeko G, Pretorius A, Walsh T, Majara L, Mndini MM, Mqulwana SG, Ntola OA, Casadei S, Ngqengelele LL, Korchina V, van der Merwe C, Malan M, Fader KM, Feng M, Willoughby E, Muzny D, Baldinger A, Andrews HF, Gur RC, Gibbs RA, Zingela Z, Nagdee M, Ramesar RS, King MC, McClellan JM. Genetics of schizophrenia in the South African Xhosa. Science 2020; 367:569-573. [PMID: 32001654 PMCID: PMC9558321 DOI: 10.1126/science.aay8833] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 12/18/2019] [Indexed: 07/20/2023]
Abstract
Africa, the ancestral home of all modern humans, is the most informative continent for understanding the human genome and its contribution to complex disease. To better understand the genetics of schizophrenia, we studied the illness in the Xhosa population of South Africa, recruiting 909 cases and 917 age-, gender-, and residence-matched controls. Individuals with schizophrenia were significantly more likely than controls to harbor private, severely damaging mutations in genes that are critical to synaptic function, including neural circuitry mediated by the neurotransmitters glutamine, γ-aminobutyric acid, and dopamine. Schizophrenia is genetically highly heterogeneous, involving severe ultrarare mutations in genes that are critical to synaptic plasticity. The depth of genetic variation in Africa revealed this relationship with a moderate sample size and informed our understanding of the genetics of schizophrenia worldwide.
Collapse
Affiliation(s)
- S Gulsuner
- Department of Medicine, Department of Genome Sciences, and Department of Psychiatry, University of Washington, Seattle, WA, USA
| | - D J Stein
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - E S Susser
- Mailman School of Public Health, Columbia University, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - G Sibeko
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - A Pretorius
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - T Walsh
- Department of Medicine, Department of Genome Sciences, and Department of Psychiatry, University of Washington, Seattle, WA, USA
| | - L Majara
- Division of Human Genetics, University of Cape Town, Cape Town, South Africa
| | - M M Mndini
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - S G Mqulwana
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - O A Ntola
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - S Casadei
- Department of Medicine, Department of Genome Sciences, and Department of Psychiatry, University of Washington, Seattle, WA, USA
| | - L L Ngqengelele
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - V Korchina
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - C van der Merwe
- Division of Human Genetics, University of Cape Town, Cape Town, South Africa
| | - M Malan
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - K M Fader
- Mailman School of Public Health, Columbia University, New York, NY, USA
| | - M Feng
- Mailman School of Public Health, Columbia University, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - E Willoughby
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - D Muzny
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - A Baldinger
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - H F Andrews
- Mailman School of Public Health, Columbia University, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - R C Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - R A Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Z Zingela
- Department of Psychology, Rhodes University, Makhanda (Grahamstown), South Africa
- Department of Psychiatry and Human Behavioral Sciences, Walter Sisulu University, Mthatha, South Africa
| | - M Nagdee
- Department of Psychology, Rhodes University, Makhanda (Grahamstown), South Africa
- Department of Psychiatry and Human Behavioral Sciences, Walter Sisulu University, Mthatha, South Africa
| | - R S Ramesar
- Division of Human Genetics, University of Cape Town, Cape Town, South Africa
| | - M-C King
- Department of Medicine, Department of Genome Sciences, and Department of Psychiatry, University of Washington, Seattle, WA, USA.
| | - J M McClellan
- Department of Medicine, Department of Genome Sciences, and Department of Psychiatry, University of Washington, Seattle, WA, USA
| |
Collapse
|
177
|
Yun Y, Hong SA, Kim KK, Baek D, Lee D, Londhe AM, Lee M, Yu J, McEachin ZT, Bassell GJ, Bowser R, Hales CM, Cho SR, Kim J, Pae AN, Cheong E, Kim S, Boulis NM, Bae S, Ha Y. CRISPR-mediated gene correction links the ATP7A M1311V mutations with amyotrophic lateral sclerosis pathogenesis in one individual. Commun Biol 2020; 3:33. [PMID: 31959876 PMCID: PMC6970999 DOI: 10.1038/s42003-020-0755-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 12/17/2019] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a severe disease causing motor neuron death, but a complete cure has not been developed and related genes have not been defined in more than 80% of cases. Here we compared whole genome sequencing results from a male ALS patient and his healthy parents to identify relevant variants, and chose one variant in the X-linked ATP7A gene, M1311V, as a strong disease-linked candidate after profound examination. Although this variant is not rare in the Ashkenazi Jewish population according to results in the genome aggregation database (gnomAD), CRISPR-mediated gene correction of this mutation in patient-derived and re-differentiated motor neurons drastically rescued neuronal activities and functions. These results suggest that the ATP7A M1311V mutation has a potential responsibility for ALS in this patient and might be a potential therapeutic target, revealed here by a personalized medicine strategy.
Collapse
Affiliation(s)
- Yeomin Yun
- Department of Neurosurgery, Spine & Spinal Cord Institute, College of Medicine, Yonsei University, Seoul, 03722, South Korea
- Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul, 03722, South Korea
| | - Sung-Ah Hong
- Department of Chemistry, Hanyang University, Seoul, 04763, South Korea
- Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, South Korea
| | - Ka-Kyung Kim
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Daye Baek
- Department of Neurosurgery, Spine & Spinal Cord Institute, College of Medicine, Yonsei University, Seoul, 03722, South Korea
- Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul, 03722, South Korea
| | - Dongsu Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, South Korea
| | - Ashwini M Londhe
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, PO Box 131, Cheongryang, Seoul, 130-650, South Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, South Korea
| | - Minhyung Lee
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, South Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, 34113, South Korea
| | - Jihyeon Yu
- Department of Chemistry, Hanyang University, Seoul, 04763, South Korea
| | - Zachary T McEachin
- Laboratory of Translational Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Gary J Bassell
- Laboratory of Translational Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Cell Biology, Emory University, Atlanta, GA, 30322, USA
| | - Robert Bowser
- Department of Neurobiology, Barrow Neurological Institute and St. Joseph's Hospital and Medical Center, Phoenix, AZ, 85013, USA
| | - Chadwick M Hales
- Department of Neurology, Emory University, Atlanta, GA, 30322, USA
| | - Sung-Rae Cho
- Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul, 03722, South Korea
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Janghwan Kim
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, South Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, 34113, South Korea
| | - Ae Nim Pae
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, PO Box 131, Cheongryang, Seoul, 130-650, South Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, South Korea
| | - Eunji Cheong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, South Korea
| | - Sangwoo Kim
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Nicholas M Boulis
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Sangsu Bae
- Department of Chemistry, Hanyang University, Seoul, 04763, South Korea.
- Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, South Korea.
| | - Yoon Ha
- Department of Neurosurgery, Spine & Spinal Cord Institute, College of Medicine, Yonsei University, Seoul, 03722, South Korea.
- Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul, 03722, South Korea.
| |
Collapse
|
178
|
Novel Approaches for Identifying the Molecular Background of Schizophrenia. Cells 2020; 9:cells9010246. [PMID: 31963710 PMCID: PMC7017322 DOI: 10.3390/cells9010246] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/06/2020] [Accepted: 01/16/2020] [Indexed: 12/20/2022] Open
Abstract
Recent advances in psychiatric genetics have led to the discovery of dozens of genomic loci associated with schizophrenia. However, a gap exists between the detection of genetic associations and understanding the underlying molecular mechanisms. This review describes the basic approaches used in the so-called post-GWAS studies to generate biological interpretation of the existing population genetic data, including both molecular (creation and analysis of knockout animals, exploration of the transcriptional effects of common variants in human brain cells) and computational (fine-mapping of causal variability, gene set enrichment analysis, partitioned heritability analysis) methods. The results of the crucial studies, in which these approaches were used to uncover the molecular and neurobiological basis of the disease, are also reported.
Collapse
|
179
|
Li M, Shen L, Chen L, Huai C, Huang H, Wu X, Yang C, Ma J, Zhou W, Du H, Fan L, He L, Wan C, Qin S. Novel genetic susceptibility loci identified by family based whole exome sequencing in Han Chinese schizophrenia patients. Transl Psychiatry 2020; 10:5. [PMID: 32066673 PMCID: PMC7026419 DOI: 10.1038/s41398-020-0708-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 12/07/2019] [Accepted: 12/19/2019] [Indexed: 12/14/2022] Open
Abstract
Schizophrenia (SCZ) is a highly heritable psychiatric disorder that affects approximately 1% of population around the world. However, early relevant studies did not reach clear conclusions of the genetic mechanisms of SCZ, suggesting that additional susceptibility loci that exert significant influence on SCZ are yet to be revealed. So, in order to identify novel susceptibility genes that account for the genetic risk of SCZ, we performed a systematic family-based study using whole exome sequencing (WES) in 65 Han Chinese families. The analysis of 51 SCZ trios with both unaffected parents identified 22 exonic and 1 splice-site de novo mutations (DNMs) on a total of 23 genes, and showed that 12 genes carried rare protein-altering compound heterozygous mutations in more than one trio. In addition, we identified 26 exonic or splice-site single nucleotide polymorphisms (SNPs) on 18 genes with nominal significance (P < 5 × 10-4) using a transmission disequilibrium test (TDT) in all the families. Moreover, TDT result confirmed a SCZ susceptibility locus on 3p21.1, encompassing the multigenetic region NEK4-ITIH1-ITIH3-ITIH4. Through several different strategies to predict the potential pathogenic genes in silico, we revealed 4 previous discovered susceptibility genes (TSNARE1, PBRM1, STAB1 and OLIG2) and 4 novel susceptibility loci (PSEN1, TLR5, MGAT5B and SSPO) in Han Chinese SCZ patients. In summary, we identified a list of putative candidate genes for SCZ using a family-based WES approach, thus improving our understanding of the pathology of SCZ and providing critical clues to future functional validation.
Collapse
Affiliation(s)
- Mo Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Lu Shen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Luan Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Cong Huai
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Hailiang Huang
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Xi Wu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Chao Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Jingsong Ma
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Wei Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Huihui Du
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Lingzi Fan
- Psychiatric Hospital of Zhumadian City, Henan, China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China.
- The Third Affiliated Hospital, Guangzhou Medical University, Guangdong, China.
| | - Chunling Wan
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China.
| | - Shengying Qin
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China.
- Collaborative Innovation Center, Jining Medical University, Shandong, China.
| |
Collapse
|
180
|
Davis BA, David F, O’Regan C, Adam MA, Harwood AJ, Crunelli V, Isles AR. Impairments in sensory-motor gating and information processing in a mouse model of Ehmt1 haploinsufficiency. Brain Neurosci Adv 2020; 4:2398212820928647. [PMID: 32954001 PMCID: PMC7479861 DOI: 10.1177/2398212820928647] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 04/30/2020] [Indexed: 12/14/2022] Open
Abstract
Regulators of chromatin dynamics and transcription are increasingly implicated in the aetiology of neurodevelopmental disorders. Haploinsufficiency of EHMT1, encoding a histone methyltransferase, is associated with several neurodevelopmental disorders, including Kleefstra syndrome, developmental delay and autism spectrum disorder. Using a mouse model of Ehmt1 haploinsufficiency (Ehmt1 D6Cre/+), we examined a number of brain and behavioural endophenotypes of relevance to neurodevelopmental disorders. Specifically, we show that Ehmt1 D6Cre/+ mice have deficits in information processing, evidenced by abnormal sensory-motor gating, a complete absence of object recognition memory, and a reduced magnitude of auditory evoked potentials in both paired-pulse inhibition and mismatch negativity. The electrophysiological experiments show that differences in magnitude response to auditory stimulus were associated with marked reductions in total and evoked beta- and gamma-band oscillatory activity, as well as significant reductions in phase synchronisation. The pattern of electrophysiological deficits in Ehmt1 D6Cre/+ matches those seen in control mice following administration of the selective NMDA-R antagonist, ketamine. This, coupled with reduction of Grin1 mRNA expression in Ehmt1 D6Cre/+ hippocampus, suggests that Ehmt1 haploinsufficiency may lead to disruption in NMDA-R. Taken together, these data indicate that reduced Ehmt1 dosage during forebrain development leads to abnormal circuitry formation, which in turn results in profound information processing deficits. Such information processing deficits are likely paramount to our understanding of the cognitive and neurological dysfunctions shared across the neurodevelopmental disorders associated with EHMT1 haploinsufficiency.
Collapse
Affiliation(s)
- Brittany A Davis
- Neuroscience and Mental Health
Research Institute and School of Biosciences, Cardiff University, Cardiff,
UK
| | - François David
- Neuroscience and Mental Health
Research Institute and School of Biosciences, Cardiff University, Cardiff,
UK
| | - Ciara O’Regan
- MRC Centre for Neuropsychiatric
Genetics and Genomics, School of Medicine, Cardiff University, Cardiff,
UK
| | - Manal A Adam
- MRC Centre for Neuropsychiatric
Genetics and Genomics, School of Medicine, Cardiff University, Cardiff,
UK
| | - Adrian J Harwood
- Neuroscience and Mental Health
Research Institute and School of Biosciences, Cardiff University, Cardiff,
UK
| | - Vincenzo Crunelli
- Neuroscience and Mental Health
Research Institute and School of Biosciences, Cardiff University, Cardiff,
UK
| | - Anthony R Isles
- MRC Centre for Neuropsychiatric
Genetics and Genomics, School of Medicine, Cardiff University, Cardiff,
UK
| |
Collapse
|
181
|
Hall LS, Medway CW, Pain O, Pardiñas AF, Rees EG, Escott-Price V, Pocklington A, Bray NJ, Holmans PA, Walters JTR, Owen MJ, O’Donovan MC. A transcriptome-wide association study implicates specific pre- and post-synaptic abnormalities in schizophrenia. Hum Mol Genet 2020; 29:159-167. [PMID: 31691811 PMCID: PMC7416679 DOI: 10.1093/hmg/ddz253] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/31/2019] [Accepted: 09/02/2019] [Indexed: 12/25/2022] Open
Abstract
Schizophrenia is a complex highly heritable disorder. Genome-wide association studies (GWAS) have identified multiple loci that influence the risk of developing schizophrenia, although the causal variants driving these associations and their impacts on specific genes are largely unknown. We identify a significant correlation between schizophrenia risk and expression at 89 genes in the dorsolateral prefrontal cortex (P ≤ 9.43 × 10-6), including 20 novel genes. Genes whose expression correlate with schizophrenia were enriched for those involved in abnormal CNS synaptic transmission (PFDR = 0.02) and antigen processing and presentation of peptide antigen via MHC class I (PFDR = 0.02). Within the CNS synaptic transmission set, we identify individual significant candidate genes to which we assign direction of expression changes in schizophrenia. The findings provide strong candidates for experimentally probing the molecular basis of synaptic pathology in schizophrenia.
Collapse
Affiliation(s)
- Lynsey S Hall
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Christopher W Medway
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Oliver Pain
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
| | - Antonio F Pardiñas
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Elliott G Rees
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Valentina Escott-Price
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Andrew Pocklington
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Nicholas J Bray
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Peter A Holmans
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - James T R Walters
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Michael J Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Michael C O’Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| |
Collapse
|
182
|
Dobbins GC, Patki A, Chen D, Tiwari HK, Hendrickson C, Britt WJ, Fowler K, Chen JY, Boppana SB, Ross SA. Association of CMV genomic mutations with symptomatic infection and hearing loss in congenital CMV infection. BMC Infect Dis 2019; 19:1046. [PMID: 31822287 PMCID: PMC6905059 DOI: 10.1186/s12879-019-4681-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/29/2019] [Indexed: 12/23/2022] Open
Abstract
Background Congenital cytomegalovirus (cCMV) infection is the most common congenital infection and a leading cause of long-term neurological and sensory sequelae, the most common being sensorineural hearing loss (SNHL). Despite extensive research, clinical or laboratory markers to identify CMV infected children with increased risk for disease have not been identified. This study utilizes viral whole-genome next generation-sequencing (NGS) of specimens from congenitally infected infants to explore viral diversity and specific viral variants that may be associated with symptomatic infection and SNHL. Methods CMV DNA from urine specimens of 30 infants (17 asymptomatic, 13 symptomatic) was target enriched and next generation sequenced resulting in 93% coverage of the CMV genome allowing analysis of viral diversity. Results Variant frequency distribution was compared between children with symptomatic and asymptomatic cCMV and those with (n = 13) and without (n = 17) hearing loss. The CMV genes UL48A, UL88, US19 and US22 were found to have an increase in nucleotide diversity in symptomatic children; while UL57, UL20, UL104, US14, UL115, and UL35 had an increase in diversity in children with hearing loss. An analysis of single variant differences between symptomatic and asymptomatic children found UL55 to have the highest number, while the most variants associated with SNHL were in the RL11 gene family. In asymptomatic infants with SNHL, mutations were observed more frequently in UL33 and UL20. Conclusion CMV genomes from infected newborns can be mapped to 93% of the genome at a depth allowing accurate and reproducible analysis of polymorphisms for variant and gene discovery that may be linked to symptomatic and hearing loss outcomes.
Collapse
Affiliation(s)
- G Clement Dobbins
- Department of Pediatrics, The University of Alabama School of Medicine, CHB 116, 1600 6th Avenue South, Birmingham, AL, USA.
| | - Amit Patki
- Department of Biostatistics, The University of Alabama School of Public Health, Birmingham, AL, USA
| | - Dongquan Chen
- Informatics Institute, The University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hemant K Tiwari
- Department of Biostatistics, The University of Alabama School of Public Health, Birmingham, AL, USA
| | - Curtis Hendrickson
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - William J Britt
- Department of Pediatrics, The University of Alabama School of Medicine, CHB 116, 1600 6th Avenue South, Birmingham, AL, USA.,Informatics Institute, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Karen Fowler
- Department of Pediatrics, The University of Alabama School of Medicine, CHB 116, 1600 6th Avenue South, Birmingham, AL, USA
| | - Jake Y Chen
- Informatics Institute, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Suresh B Boppana
- Department of Pediatrics, The University of Alabama School of Medicine, CHB 116, 1600 6th Avenue South, Birmingham, AL, USA.,Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shannon A Ross
- Department of Pediatrics, The University of Alabama School of Medicine, CHB 116, 1600 6th Avenue South, Birmingham, AL, USA. .,Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
183
|
Farhan SMK, Howrigan DP, Abbott LE, Klim JR, Topp SD, Byrnes AE, Churchhouse C, Phatnani H, Smith BN, Rampersaud E, Wu G, Wuu J, Shatunov A, Iacoangeli A, Al Khleifat A, Mordes DA, Ghosh S, Eggan K, Rademakers R, McCauley JL, Schüle R, Züchner S, Benatar M, Taylor JP, Nalls M, Gotkine M, Shaw PJ, Morrison KE, Al-Chalabi A, Traynor B, Shaw CE, Goldstein DB, Harms MB, Daly MJ, Neale BM. Exome sequencing in amyotrophic lateral sclerosis implicates a novel gene, DNAJC7, encoding a heat-shock protein. Nat Neurosci 2019; 22:1966-1974. [PMID: 31768050 PMCID: PMC6919277 DOI: 10.1038/s41593-019-0530-0] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 10/02/2019] [Indexed: 12/11/2022]
Abstract
To discover novel genes underlying amyotrophic lateral sclerosis (ALS), we aggregated exomes from 3,864 cases and 7,839 ancestry-matched controls. We observed a significant excess of rare protein-truncating variants among ALS cases, and these variants were concentrated in constrained genes. Through gene level analyses, we replicated known ALS genes including SOD1, NEK1 and FUS. We also observed multiple distinct protein-truncating variants in a highly constrained gene, DNAJC7. The signal in DNAJC7 exceeded genome-wide significance, and immunoblotting assays showed depletion of DNAJC7 protein in fibroblasts in a patient with ALS carrying the p.Arg156Ter variant. DNAJC7 encodes a member of the heat-shock protein family, HSP40, which, along with HSP70 proteins, facilitates protein homeostasis, including folding of newly synthesized polypeptides and clearance of degraded proteins. When these processes are not regulated, misfolding and accumulation of aberrant proteins can occur and lead to protein aggregation, which is a pathological hallmark of neurodegeneration. Our results highlight DNAJC7 as a novel gene for ALS.
Collapse
Affiliation(s)
- Sali M K Farhan
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| | - Daniel P Howrigan
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Liam E Abbott
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Joseph R Klim
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Simon D Topp
- United Kingdom Dementia Research Institute Centre, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Andrea E Byrnes
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Claire Churchhouse
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Hemali Phatnani
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY, USA
| | - Bradley N Smith
- United Kingdom Dementia Research Institute Centre, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Evadnie Rampersaud
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Gang Wu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Joanne Wuu
- Department of Neurology, University of Miami, Miami, FL, USA
| | - Aleksey Shatunov
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
| | - Alfredo Iacoangeli
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Ahmad Al Khleifat
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
| | - Daniel A Mordes
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Sulagna Ghosh
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Kevin Eggan
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Jacob L McCauley
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, USA
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Rebecca Schüle
- Center for Neurology and Hertie Institute für Clinical Brain Research, University of Tübingen, German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Stephan Züchner
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, USA
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Michael Benatar
- Department of Neurology, University of Miami, Miami, FL, USA
| | - J Paul Taylor
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Michael Nalls
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Data Tecnica International, Glen Echo, MD, USA
| | - Marc Gotkine
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Karen E Morrison
- Faculty of Medicine, University of Southampton and Department of Neurology, University Hospital Southampton, Southampton, UK
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
- Department of Neurology, King's College Hospital, London, UK
| | - Bryan Traynor
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Christopher E Shaw
- United Kingdom Dementia Research Institute Centre, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - David B Goldstein
- Institute for Genomic Medicine, Columbia University, New York, NY, USA
| | - Matthew B Harms
- Department of Neurology, Columbia University, New York, NY, USA
| | - Mark J Daly
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Benjamin M Neale
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
184
|
Abstract
PURPOSE OF REVIEW To summarize the molecular and clinical findings of KMT2B-related dystonia (DYT-KMT2B), a newly identified genetic dystonia syndrome. RECENT FINDINGS Since first described in 2016, 66 different KMT2B-affecting variants, encompassing a set of frameshift, nonsense, splice-site, missense, and deletion mutations, have been reported in 76 patients. Most mutations are de novo and expected to mediate epigenetic dysregulation by inducing KMT2B haploinsufficiency. DYT-KMT2B is characterized phenotypically by limb-onset childhood dystonia that tends to spread progressively, resulting in generalized dystonia with cranio-cervical involvement. Co-occuring signs such as intellectual disability are frequently observed. Sustained response to deep brain stimulation (DBS), including restoration of independent ambulation, is seen in 93% (27/29) of patients. DYT-KMT2B is emerging as a prevalent monogenic dystonia. Childhood-onset dystonia presentations should prompt a search for KMT2B mutations, preferentially via next-generation-sequencing and genomic-array technologies, to enable specific counseling and treatment. Prospective multicenter studies are desirable to establish KMT2B mutational status as a DBS outcome predictor.
Collapse
Affiliation(s)
- Michael Zech
- Institut für Neurogenomik, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Munich, Neuherberg, Germany.,Institut für Humangenetik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Daniel D Lam
- Institut für Neurogenomik, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Munich, Neuherberg, Germany
| | - Juliane Winkelmann
- Institut für Neurogenomik, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Munich, Neuherberg, Germany. .,Institut für Humangenetik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany. .,Lehrstuhl für Neurogenetik, Technische Universität München, Munich, Germany. .,Munich Cluster for Systems Neurology, SyNergy, Munich, Germany.
| |
Collapse
|
185
|
van 't Hof FNG, Lai D, van Setten J, Bots ML, Vaartjes I, Broderick J, Woo D, Foroud T, Rinkel GJE, de Bakker PIW, Ruigrok YM. Exome-chip association analysis of intracranial aneurysms. Neurology 2019; 94:e481-e488. [PMID: 31732565 DOI: 10.1212/wnl.0000000000008665] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 08/01/2019] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To investigate to what extent low-frequency genetic variants (with minor allele frequencies <5%) affect the risk of intracranial aneurysms (IAs). METHODS One thousand fifty-six patients with IA and 2,097 population-based controls from the Netherlands were genotyped with the Illumina HumanExome BeadChip. After quality control (QC) of samples and single nucleotide variants (SNVs), we conducted a single variant analysis using the Fisher exact test. We also performed the variable threshold (VT) test and the sequence kernel association test (SKAT) at different minor allele count (MAC) thresholds of >5 and >0 to test the hypothesis that multiple variants within the same gene are associated with IA risk. Significant results were tested in a replication cohort of 425 patients with IA and 311 controls, and results of the 2 cohorts were combined in a meta-analysis. RESULTS After QC, 995 patients with IA and 2,080 controls remained for further analysis. The single variant analysis comprising 46,534 SNVs did not identify significant loci at the genome-wide level. The gene-based tests showed a statistically significant association for fibulin 2 (FBLN2) (best p = 1 × 10-6 for the VT test, MAC >5). Associations were not statistically significant in the independent but smaller replication cohort (p > 0.57) but became slightly stronger in a meta-analysis of the 2 cohorts (best p = 4.8 × 10-7 for the SKAT, MAC ≥1). CONCLUSION Gene-based tests indicated an association for FBLN2, a gene encoding an extracellular matrix protein implicated in vascular wall remodeling, but independent validation in larger cohorts is warranted. We did not identify any significant associations for single low-frequency genetic variants.
Collapse
Affiliation(s)
- Femke N G van 't Hof
- From the Department of Neurology and Neurosurgery (F.N.G.v.H., G.J.E.R., Y.M.R.), Brain Center Rudolf Magnus, Department of Cardiology (J.v.S.), Department of Medical Genetics (P.I.W.d.B.), Centre for Molecular Medicine, and Department of Epidemiology (M.L.B., I.V., P.I.W.d.B.), Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, the Netherlands; Department of Medical and Molecular Genetics (D.L., T.F.), Indiana University School of Medicine, Indianapolis; and Department of Neurology and Rehabilitation Medicine (J.B., D.W.), University of Cincinnati School of Medicine, OH.
| | - Dongbing Lai
- From the Department of Neurology and Neurosurgery (F.N.G.v.H., G.J.E.R., Y.M.R.), Brain Center Rudolf Magnus, Department of Cardiology (J.v.S.), Department of Medical Genetics (P.I.W.d.B.), Centre for Molecular Medicine, and Department of Epidemiology (M.L.B., I.V., P.I.W.d.B.), Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, the Netherlands; Department of Medical and Molecular Genetics (D.L., T.F.), Indiana University School of Medicine, Indianapolis; and Department of Neurology and Rehabilitation Medicine (J.B., D.W.), University of Cincinnati School of Medicine, OH
| | - Jessica van Setten
- From the Department of Neurology and Neurosurgery (F.N.G.v.H., G.J.E.R., Y.M.R.), Brain Center Rudolf Magnus, Department of Cardiology (J.v.S.), Department of Medical Genetics (P.I.W.d.B.), Centre for Molecular Medicine, and Department of Epidemiology (M.L.B., I.V., P.I.W.d.B.), Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, the Netherlands; Department of Medical and Molecular Genetics (D.L., T.F.), Indiana University School of Medicine, Indianapolis; and Department of Neurology and Rehabilitation Medicine (J.B., D.W.), University of Cincinnati School of Medicine, OH
| | - Michiel L Bots
- From the Department of Neurology and Neurosurgery (F.N.G.v.H., G.J.E.R., Y.M.R.), Brain Center Rudolf Magnus, Department of Cardiology (J.v.S.), Department of Medical Genetics (P.I.W.d.B.), Centre for Molecular Medicine, and Department of Epidemiology (M.L.B., I.V., P.I.W.d.B.), Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, the Netherlands; Department of Medical and Molecular Genetics (D.L., T.F.), Indiana University School of Medicine, Indianapolis; and Department of Neurology and Rehabilitation Medicine (J.B., D.W.), University of Cincinnati School of Medicine, OH
| | - Ilonca Vaartjes
- From the Department of Neurology and Neurosurgery (F.N.G.v.H., G.J.E.R., Y.M.R.), Brain Center Rudolf Magnus, Department of Cardiology (J.v.S.), Department of Medical Genetics (P.I.W.d.B.), Centre for Molecular Medicine, and Department of Epidemiology (M.L.B., I.V., P.I.W.d.B.), Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, the Netherlands; Department of Medical and Molecular Genetics (D.L., T.F.), Indiana University School of Medicine, Indianapolis; and Department of Neurology and Rehabilitation Medicine (J.B., D.W.), University of Cincinnati School of Medicine, OH
| | - Joseph Broderick
- From the Department of Neurology and Neurosurgery (F.N.G.v.H., G.J.E.R., Y.M.R.), Brain Center Rudolf Magnus, Department of Cardiology (J.v.S.), Department of Medical Genetics (P.I.W.d.B.), Centre for Molecular Medicine, and Department of Epidemiology (M.L.B., I.V., P.I.W.d.B.), Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, the Netherlands; Department of Medical and Molecular Genetics (D.L., T.F.), Indiana University School of Medicine, Indianapolis; and Department of Neurology and Rehabilitation Medicine (J.B., D.W.), University of Cincinnati School of Medicine, OH
| | - Daniel Woo
- From the Department of Neurology and Neurosurgery (F.N.G.v.H., G.J.E.R., Y.M.R.), Brain Center Rudolf Magnus, Department of Cardiology (J.v.S.), Department of Medical Genetics (P.I.W.d.B.), Centre for Molecular Medicine, and Department of Epidemiology (M.L.B., I.V., P.I.W.d.B.), Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, the Netherlands; Department of Medical and Molecular Genetics (D.L., T.F.), Indiana University School of Medicine, Indianapolis; and Department of Neurology and Rehabilitation Medicine (J.B., D.W.), University of Cincinnati School of Medicine, OH
| | - Tatiana Foroud
- From the Department of Neurology and Neurosurgery (F.N.G.v.H., G.J.E.R., Y.M.R.), Brain Center Rudolf Magnus, Department of Cardiology (J.v.S.), Department of Medical Genetics (P.I.W.d.B.), Centre for Molecular Medicine, and Department of Epidemiology (M.L.B., I.V., P.I.W.d.B.), Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, the Netherlands; Department of Medical and Molecular Genetics (D.L., T.F.), Indiana University School of Medicine, Indianapolis; and Department of Neurology and Rehabilitation Medicine (J.B., D.W.), University of Cincinnati School of Medicine, OH
| | - Gabriel J E Rinkel
- From the Department of Neurology and Neurosurgery (F.N.G.v.H., G.J.E.R., Y.M.R.), Brain Center Rudolf Magnus, Department of Cardiology (J.v.S.), Department of Medical Genetics (P.I.W.d.B.), Centre for Molecular Medicine, and Department of Epidemiology (M.L.B., I.V., P.I.W.d.B.), Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, the Netherlands; Department of Medical and Molecular Genetics (D.L., T.F.), Indiana University School of Medicine, Indianapolis; and Department of Neurology and Rehabilitation Medicine (J.B., D.W.), University of Cincinnati School of Medicine, OH
| | - Paul I W de Bakker
- From the Department of Neurology and Neurosurgery (F.N.G.v.H., G.J.E.R., Y.M.R.), Brain Center Rudolf Magnus, Department of Cardiology (J.v.S.), Department of Medical Genetics (P.I.W.d.B.), Centre for Molecular Medicine, and Department of Epidemiology (M.L.B., I.V., P.I.W.d.B.), Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, the Netherlands; Department of Medical and Molecular Genetics (D.L., T.F.), Indiana University School of Medicine, Indianapolis; and Department of Neurology and Rehabilitation Medicine (J.B., D.W.), University of Cincinnati School of Medicine, OH
| | - Ynte M Ruigrok
- From the Department of Neurology and Neurosurgery (F.N.G.v.H., G.J.E.R., Y.M.R.), Brain Center Rudolf Magnus, Department of Cardiology (J.v.S.), Department of Medical Genetics (P.I.W.d.B.), Centre for Molecular Medicine, and Department of Epidemiology (M.L.B., I.V., P.I.W.d.B.), Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, the Netherlands; Department of Medical and Molecular Genetics (D.L., T.F.), Indiana University School of Medicine, Indianapolis; and Department of Neurology and Rehabilitation Medicine (J.B., D.W.), University of Cincinnati School of Medicine, OH
| |
Collapse
|
186
|
Cyrus S, Burkardt D, Weaver DD, Gibson WT. PRC2-complex related dysfunction in overgrowth syndromes: A review of EZH2, EED, and SUZ12 and their syndromic phenotypes. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2019; 181:519-531. [PMID: 31724824 DOI: 10.1002/ajmg.c.31754] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 10/18/2019] [Accepted: 10/23/2019] [Indexed: 12/31/2022]
Abstract
The EZH2, EED, and SUZ12 genes encode proteins that comprise core components of the polycomb repressive complex 2 (PRC2), an epigenetic "writer" with H3K27 methyltransferase activity, catalyzing the addition of up to three methyl groups on histone 3 at lysine residue 27 (H3K27). Partial loss-of-function variants in genes encoding the EZH2 and EED subunits of the complex lead to overgrowth, macrocephaly, advanced bone age, variable intellectual disability, and distinctive facial features. EZH2-associated overgrowth, caused by constitutional heterozygous mutations within Enhancer of Zeste homologue 2 (EZH2), has a phenotypic spectrum ranging from tall stature without obvious intellectual disability or dysmorphic features to classical Weaver syndrome (OMIM #277590). EED-associated overgrowth (Cohen-Gibson syndrome; OMIM #617561) is caused by germline heterozygous mutations in Embryonic Ectoderm Development (EED), and manifests overgrowth and intellectual disability (OGID), along with other features similar to Weaver syndrome. Most recently, rare coding variants in SUZ12 have also been described that present with clinical characteristics similar to the previous two syndromes. Here we review the PRC2 complex and clinical syndromes of OGID associated with core components EZH2, EED, and SUZ12.
Collapse
Affiliation(s)
- Sharri Cyrus
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Deepika Burkardt
- Center for Human Genetics, University Hospitals Rainbow Babies and Children/Department of Genetics, Case Western Reserve University, Cleveland, Ohio
| | - David D Weaver
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - William T Gibson
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.,British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
187
|
Escott-Price V, Smith DJ, Kendall K, Ward J, Kirov G, Owen MJ, Walters J, O’Donovan MC. Polygenic risk for schizophrenia and season of birth within the UK Biobank cohort. Psychol Med 2019; 49:2499-2504. [PMID: 29501066 PMCID: PMC7610956 DOI: 10.1017/s0033291718000454] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND There is strong evidence that people born in winter and in spring have a small increased risk of schizophrenia. As this 'season of birth' effect underpins some of the most influential hypotheses concerning potentially modifiable risk exposures, it is important to exclude other possible explanations for the phenomenon. METHODS Here we sought to determine whether the season of birth effect reflects gene-environment confounding rather than a pathogenic process indexing environmental exposure. We directly measured, in 136 538 participants from the UK Biobank (UKBB), the burdens of common schizophrenia risk alleles and of copy number variants known to increase the risk for the disorder, and tested whether these were correlated with a season of birth. RESULTS Neither genetic measure was associated with season or month of birth within the UKBB sample. CONCLUSIONS As our study was highly powered to detect small effects, we conclude that the season of birth effect in schizophrenia reflects a true pathogenic effect of environmental exposure.
Collapse
Affiliation(s)
| | - Daniel J. Smith
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Kimberley Kendall
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Joey Ward
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - George Kirov
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Michael J. Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - James Walters
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Michael C. O’Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| |
Collapse
|
188
|
Alexander Arguello P, Addington A, Borja S, Brady L, Dutka T, Gitik M, Koester S, Meinecke D, Merikangas K, McMahon FJ, Panchision D, Senthil G, Lehner T. From genetics to biology: advancing mental health research in the Genomics ERA. Mol Psychiatry 2019; 24:1576-1582. [PMID: 31164699 DOI: 10.1038/s41380-019-0445-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 11/09/2022]
Abstract
The Genomics Workgroup of the National Advisory Mental Health Council (NAMHC) recently issued a set of recommendations for advancing the NIMH psychiatric genetics research program and prioritizing subsequent follow-up studies. The report emphasized the primacy of rigorous statistical support from properly designed, well-powered studies for pursuing genetic variants robustly associated with disease. Here we discuss the major points NIMH program staff consider when assessing research applications based on common and rare variants, as well as genetic syndromes, associated with psychiatric disorders. These are broad guiding principles for investigators to consider prior to submission of their applications. NIMH staff weigh these points in the context of reviewer comments, the existing literature, and current investments in related projects. Following the recommendations of the NAMHC, statistical strength and robustness of the underlying genetic discovery weighs heavily in our funding considerations as does the suitability of the proposed experimental approach. We specifically address our evaluation of applications motivated in whole, or in part, by an association between human DNA sequence variation and a disease or trait relevant to the mission of the NIMH.
Collapse
Affiliation(s)
- P Alexander Arguello
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Anjené Addington
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Susan Borja
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Linda Brady
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Tara Dutka
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Miri Gitik
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Susan Koester
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Douglas Meinecke
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Kathleen Merikangas
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Francis J McMahon
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - David Panchision
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Geetha Senthil
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Thomas Lehner
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
189
|
Qi G, Han C, Sun Y, Zhou Y. Genetic insight into cytochrome P450 in Chinese from the Chinese Millionome Database. Basic Clin Pharmacol Toxicol 2019; 126:341-352. [PMID: 31661191 DOI: 10.1111/bcpt.13356] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 10/15/2019] [Indexed: 12/21/2022]
Abstract
Genetic variations of cytochrome P450 (CYP) influence the inter-individual differences in drug response. Here, we collected 8682 variants of 57 CYP genes and cytochrome P450 oxidoreductase (POR) from a large-scale sequencing project in Chinese, Chinese Millionome Database (CMDB). In addition, 52 294 variants from the Genome Aggregation Database (gnomAD) had been simultaneously identified and analysed. Rare variants with a variant allele frequency (VAF) < 0.01 comprised 41.4% (3594/8682) of identified variations in the CMDB, while 98.1% (51 320/52 294) in the gnomAD were rare. Out of 8682 variants in the CMDB, 66.9% (5808/8682) were in introns and only 4.3% (377/8682) were missense variants. In contrast, 36.2% (18 929/52 294) variants in the gnomAD were missense. The common alleles with a VAF over 0.1 were found in CYP1A2*1C, CYP1A2*1F, CYP2C19*2, CYP2D6*2, CYP2D6*10, CYP3A5*3 and CYP4F2*3, with a VAF of 0.161, 0.6, 0.27, 0.274, 0.678, 0.92 and 0.233, respectively. The growing number of genetic variations in CYP genes as more genomes are sequenced would increase the power to predict drug metabolism and response based on the genotype of the particular individual.
Collapse
Affiliation(s)
- Guangzhao Qi
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chao Han
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ya Sun
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yubing Zhou
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
190
|
Shohat S, Shifman S. Genes essential for embryonic stem cells are associated with neurodevelopmental disorders. Genome Res 2019; 29:1910-1918. [PMID: 31649057 PMCID: PMC6836742 DOI: 10.1101/gr.250019.119] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 10/01/2019] [Indexed: 12/18/2022]
Abstract
Mouse embryonic stem cells (mESCs) are key components in generating mouse models for human diseases and performing basic research on pluripotency, yet the number of genes essential for mESCs is still unknown. We performed a genome-wide screen for essential genes in mESCs and compared it to screens in human cells. We found that essential genes are enriched for basic cellular functions, are highly expressed in mESCs, and tend to lack paralog genes. We discovered that genes that are essential specifically in mESCs play a role in pathways associated with their pluripotent state. We show that 29.5% of human genes intolerant to loss-of-function mutations are essential in mouse or human ESCs, and that the human phenotypes most significantly associated with genes essential for ESCs are neurodevelopmental. Our results provide insights into essential genes in the mouse, the pathways which govern pluripotency, and suggest that many genes associated with neurodevelopmental disorders are essential at very early embryonic stages.
Collapse
Affiliation(s)
- Shahar Shohat
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Sagiv Shifman
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| |
Collapse
|
191
|
Mukai J, Cannavò E, Crabtree GW, Sun Z, Diamantopoulou A, Thakur P, Chang CY, Cai Y, Lomvardas S, Takata A, Xu B, Gogos JA. Recapitulation and Reversal of Schizophrenia-Related Phenotypes in Setd1a-Deficient Mice. Neuron 2019; 104:471-487.e12. [PMID: 31606247 DOI: 10.1016/j.neuron.2019.09.014] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 07/28/2019] [Accepted: 09/10/2019] [Indexed: 12/15/2022]
Abstract
SETD1A, a lysine-methyltransferase, is a key schizophrenia susceptibility gene. Mice carrying a heterozygous loss-of-function mutation of the orthologous gene exhibit alterations in axonal branching and cortical synaptic dynamics accompanied by working memory deficits. We show that Setd1a binds both promoters and enhancers with a striking overlap between Setd1a and Mef2 on enhancers. Setd1a targets are highly expressed in pyramidal neurons and display a complex pattern of transcriptional up- and downregulations shaped by presumed opposing functions of Setd1a on promoters and Mef2-bound enhancers. Notably, evolutionarily conserved Setd1a targets are associated with neuropsychiatric genetic risk burden. Reinstating Setd1a expression in adulthood rescues cognitive deficits. Finally, we identify LSD1 as a major counteracting demethylase for Setd1a and show that its pharmacological antagonism results in a full rescue of the behavioral and morphological deficits in Setd1a-deficient mice. Our findings advance understanding of how SETD1A mutations predispose to schizophrenia (SCZ) and point to novel therapeutic interventions.
Collapse
Affiliation(s)
- Jun Mukai
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027, USA
| | - Enrico Cannavò
- Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027, USA
| | - Gregg W Crabtree
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027, USA
| | - Ziyi Sun
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Anastasia Diamantopoulou
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Pratibha Thakur
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027, USA
| | - Chia-Yuan Chang
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Yifei Cai
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Stavros Lomvardas
- Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027, USA; Department of Biochemistry and Molecular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Department of Neuroscience, Columbia University, New York, NY 10032, USA
| | - Atsushi Takata
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Fukuura 3-9, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Bin Xu
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA
| | - Joseph A Gogos
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027, USA; Department of Neuroscience, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
192
|
Sanders SJ, Sahin M, Hostyk J, Thurm A, Jacquemont S, Avillach P, Douard E, Martin CL, Modi ME, Moreno-De-Luca A, Raznahan A, Anticevic A, Dolmetsch R, Feng G, Geschwind DH, Glahn DC, Goldstein DB, Ledbetter DH, Mulle JG, Pasca SP, Samaco R, Sebat J, Pariser A, Lehner T, Gur RE, Bearden CE. A framework for the investigation of rare genetic disorders in neuropsychiatry. Nat Med 2019; 25:1477-1487. [PMID: 31548702 PMCID: PMC8656349 DOI: 10.1038/s41591-019-0581-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 07/31/2019] [Indexed: 02/07/2023]
Abstract
De novo and inherited rare genetic disorders (RGDs) are a major cause of human morbidity, frequently involving neuropsychiatric symptoms. Recent advances in genomic technologies and data sharing have revolutionized the identification and diagnosis of RGDs, presenting an opportunity to elucidate the mechanisms underlying neuropsychiatric disorders by investigating the pathophysiology of high-penetrance genetic risk factors. Here we seek out the best path forward for achieving these goals. We think future research will require consistent approaches across multiple RGDs and developmental stages, involving both the characterization of shared neuropsychiatric dimensions in humans and the identification of neurobiological commonalities in model systems. A coordinated and concerted effort across patients, families, researchers, clinicians and institutions, including rapid and broad sharing of data, is now needed to translate these discoveries into urgently needed therapies.
Collapse
Affiliation(s)
- Stephan J Sanders
- Department of Psychiatry, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Mustafa Sahin
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Joseph Hostyk
- Institute for Genomic Medicine, Columbia University Medical Center, Hammer Health Sciences, New York, NY, USA
| | - Audrey Thurm
- National Institute of Mental Health, Bethesda, MD, USA
| | - Sebastien Jacquemont
- CHU Sainte-Justine Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Paul Avillach
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Elise Douard
- CHU Sainte-Justine Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Christa L Martin
- Geisinger Autism & Developmental Medicine Institute, Danville, PA, USA
| | - Meera E Modi
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | - Alan Anticevic
- Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Ricardo Dolmetsch
- Department of Neuroscience, Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Guoping Feng
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Daniel H Geschwind
- Center for Autism Research and Treatment, Semel Institute for Neuroscience and Human Behavior and Departments of Neurology and Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - David C Glahn
- Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston, MA, USA
| | - David B Goldstein
- Institute for Genomic Medicine, Columbia University Medical Center, Hammer Health Sciences, New York, NY, USA
| | - David H Ledbetter
- Geisinger Autism & Developmental Medicine Institute, Danville, PA, USA
| | - Jennifer G Mulle
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Sergiu P Pasca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, CA, USA
| | - Rodney Samaco
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jonathan Sebat
- Beyster Center for Genomics of Psychiatric Diseases, University of California, San Diego, La Jolla, CA, USA
| | - Anne Pariser
- National Center for Advancing Translational Sciences, Bethesda, MD, USA
| | - Thomas Lehner
- National Institute of Mental Health, Bethesda, MD, USA
| | - Raquel E Gur
- Department of Psychiatry, Neuropsychiatry Section, and the Lifespan Brain Institute, Perelman School of Medicine and Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA.
| | - Carrie E Bearden
- Semel Institute for Neuroscience and Human Behavior, Departments of Psychiatry and Biobehavioral Sciences and Psychology, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
193
|
Duan J, Sanders AR, Gejman PV. From Schizophrenia Genetics to Disease Biology: Harnessing New Concepts and Technologies. JOURNAL OF PSYCHIATRY AND BRAIN SCIENCE 2019; 4:e190014. [PMID: 31555746 PMCID: PMC6760308 DOI: 10.20900/jpbs.20190014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Schizophrenia (SZ) is a severe mental disorder afflicting around 1% of the population. It is highly heritable but with complex genetics. Recent research has unraveled a plethora of risk loci for SZ. Accordingly, our conceptual understanding of SZ genetics has been rapidly evolving, from oligogenic models towards polygenic or even omnigenic models. A pressing challenge to the field, however, is the translation of the many genetic findings of SZ into disease biology insights leading to more effective treatments. Bridging this gap requires the integration of genetic findings and functional genomics using appropriate cellular models. Harnessing new technologies, such as the development of human induced pluripotent stem cells (hiPSC) and the CRISPR/Cas-based genome/epigenome editing approach are expected to change our understanding of SZ disease biology to a fundamentally higher level. Here, we discuss some new developments.
Collapse
Affiliation(s)
- Jubao Duan
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
- Department of Psychiatry and Behavioral Neurosciences, The University of Chicago, Chicago, IL 60637, USA
| | - Alan R. Sanders
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
- Department of Psychiatry and Behavioral Neurosciences, The University of Chicago, Chicago, IL 60637, USA
| | - Pablo V. Gejman
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
- Department of Psychiatry and Behavioral Neurosciences, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
194
|
Doostparast Torshizi A, Armoskus C, Zhang H, Forrest MP, Zhang S, Souaiaia T, Evgrafov OV, Knowles JA, Duan J, Wang K. Deconvolution of transcriptional networks identifies TCF4 as a master regulator in schizophrenia. SCIENCE ADVANCES 2019; 5:eaau4139. [PMID: 31535015 PMCID: PMC6739105 DOI: 10.1126/sciadv.aau4139] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/08/2019] [Indexed: 06/10/2023]
Abstract
Applying tissue-specific deconvolution of transcriptional networks to identify their master regulators (MRs) in neuropsychiatric disorders has been largely unexplored. Here, using two schizophrenia (SCZ) case-control RNA-seq datasets, one on postmortem dorsolateral prefrontal cortex (DLPFC) and another on cultured olfactory neuroepithelium, we deconvolved the transcriptional networks and identified TCF4 as a top candidate MR that may be dysregulated in SCZ. We validated TCF4 as a MR through enrichment analysis of TCF4-binding sites in induced pluripotent stem cell (hiPSC)-derived neurons and in neuroblastoma cells. We further validated the predicted TCF4 targets by knocking down TCF4 in hiPSC-derived neural progenitor cells (NPCs) and glutamatergic neurons (Glut_Ns). The perturbed TCF4 gene network in NPCs was more enriched for pathways involved in neuronal activity and SCZ-associated risk genes, compared to Glut_Ns. Our results suggest that TCF4 may serve as a MR of a gene network dysregulated in SCZ at early stages of neurodevelopment.
Collapse
Affiliation(s)
- Abolfazl Doostparast Torshizi
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chris Armoskus
- College of Medicine, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
- Zilkhe Neurogenetic Institute, University of Southern California, Los Angeles, CA 90089, USA
| | - Hanwen Zhang
- Center for Psychiatric Genetics, North Shore University Health System, Evanston, IL 60201, USA
| | - Marc P. Forrest
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Center for Autism and Neurodevelopment, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Siwei Zhang
- Center for Psychiatric Genetics, North Shore University Health System, Evanston, IL 60201, USA
- Department of Psychiatry and Behavioral Neurosciences, University of Chicago, Chicago, IL 60015, USA
| | - Tade Souaiaia
- College of Medicine, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
- Zilkhe Neurogenetic Institute, University of Southern California, Los Angeles, CA 90089, USA
| | - Oleg V. Evgrafov
- College of Medicine, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
- Zilkhe Neurogenetic Institute, University of Southern California, Los Angeles, CA 90089, USA
| | - James A. Knowles
- College of Medicine, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
- Zilkhe Neurogenetic Institute, University of Southern California, Los Angeles, CA 90089, USA
| | - Jubao Duan
- Center for Psychiatric Genetics, North Shore University Health System, Evanston, IL 60201, USA
- Department of Psychiatry and Behavioral Neurosciences, University of Chicago, Chicago, IL 60015, USA
| | - Kai Wang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Zilkhe Neurogenetic Institute, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
195
|
Affiliation(s)
- Jordan W Smoller
- Department of Psychiatry and Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston; and Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass
| |
Collapse
|
196
|
Post-translational histone modifications and their interaction with sex influence normal brain development and elaboration of neuropsychiatric disorders. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1968-1981. [DOI: 10.1016/j.bbadis.2018.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 02/06/2023]
|
197
|
Escott-Price V, Pardiñas AF, Santiago E, Walters J, Kirov G, Owen MJ, O'Donovan MC. The Relationship Between Common Variant Schizophrenia Liability and Number of Offspring in the UK Biobank. Am J Psychiatry 2019; 176:661-666. [PMID: 30606050 DOI: 10.1176/appi.ajp.2018.18020140] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Schizophrenia is associated with a marked reduction in reproductive success, yet alleles that are common contribute substantially to the liability of the disorder. Among several possible explanations for this, it has been postulated that individuals who carry risk alleles but are unaffected are at some reproductive advantage, offsetting the effects of negative selection among those who are affected. The authors sought to test this hypothesis, isolating the effects of risk alleles on fecundity from the effects that are contingent on expressing schizophrenia. METHODS The burden of schizophrenia risk alleles, as indexed by a polygenic risk score (PRS), carried by 139,679 participants in the UK Biobank study who did not have schizophrenia was compared with the number of offspring of these individuals. RESULTS Higher schizophrenia liability in study subjects without manifest disorder was weakly but significantly associated with having more children (B=0.006, 95% CI=0.002, 0.010). The relationship was dependent on sex, with a positive correlation between number of children and liability among females (B=0.011, 95% CI=0.006, 0.016), whereas among males, higher liability was associated with being childless (odds ratio=0.96, 95% CI=0.94, 0.98). The negative effect on number of children associated with schizophrenia itself was twofold to 15-fold greater than the positive effect associated with PRS in unaffected individuals. CONCLUSIONS These findings suggest that a complex relationship between liability and fecundity is consistent with sexual selection. Although the overall pattern of a weak positive correlation with liability may contribute to the persistence of schizophrenia risk alleles, these results indicate that the negative selection acting on individuals affected by schizophrenia in the general population is larger than any advantage conferred by genetic loading in unaffected individuals.
Collapse
Affiliation(s)
- Valentina Escott-Price
- From the MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Wales, United Kingdom (Escott-Price, Pardiñas, Walters, Kirov, Owen, O'Donovan); and the Department of Functional Biology, Faculty of Biology, University of Oviedo, Oviedo, Spain (Santiago)
| | - Antonio F Pardiñas
- From the MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Wales, United Kingdom (Escott-Price, Pardiñas, Walters, Kirov, Owen, O'Donovan); and the Department of Functional Biology, Faculty of Biology, University of Oviedo, Oviedo, Spain (Santiago)
| | - Enrique Santiago
- From the MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Wales, United Kingdom (Escott-Price, Pardiñas, Walters, Kirov, Owen, O'Donovan); and the Department of Functional Biology, Faculty of Biology, University of Oviedo, Oviedo, Spain (Santiago)
| | - James Walters
- From the MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Wales, United Kingdom (Escott-Price, Pardiñas, Walters, Kirov, Owen, O'Donovan); and the Department of Functional Biology, Faculty of Biology, University of Oviedo, Oviedo, Spain (Santiago)
| | - George Kirov
- From the MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Wales, United Kingdom (Escott-Price, Pardiñas, Walters, Kirov, Owen, O'Donovan); and the Department of Functional Biology, Faculty of Biology, University of Oviedo, Oviedo, Spain (Santiago)
| | - Michael J Owen
- From the MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Wales, United Kingdom (Escott-Price, Pardiñas, Walters, Kirov, Owen, O'Donovan); and the Department of Functional Biology, Faculty of Biology, University of Oviedo, Oviedo, Spain (Santiago)
| | - Michael C O'Donovan
- From the MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Wales, United Kingdom (Escott-Price, Pardiñas, Walters, Kirov, Owen, O'Donovan); and the Department of Functional Biology, Faculty of Biology, University of Oviedo, Oviedo, Spain (Santiago)
| |
Collapse
|
198
|
Abstract
Zusammenfassung
Häufige Krankheiten, die sog. Volkskrankheiten, sind in der Regel multifaktoriell verursacht, d. h. zu ihrer Entwicklung tragen sowohl genetische Faktoren als auch nicht-genetische Umgebungseinflüsse bei. Die geschätzte Gesamterblichkeit (‑heritabilität) reicht von moderat bis vergleichsweise hoch. Die genetische Architektur ist komplex und kann das gesamte allelische Spektrum, von häufigen Varianten mit niedriger Penetranz bis hin zu seltenen Varianten mit höherer Penetranz, sowie alle möglichen Kombinationen umfassen. Während häufige Varianten seit mehreren Jahren mit großem Erfolg durch genomweite Assoziationsstudien (GWAS) identifiziert werden, war bisher die Identifizierung seltener Varianten, insbesondere aufgrund der großen Zahl beitragender Gene, nur begrenzt erfolgreich. Dies ändert sich derzeit dank der Anwendung von Hochdurchsatz-Sequenziertechnologien („next-generation sequencing“, NGS) und der daraus resultierenden zunehmenden Verfügbarkeit von exom- und genomweiten Sequenzdaten großer Kollektive. In diesem Artikel geben wir einen Überblick über die Bedeutung seltener Varianten bei häufigen Erkrankungen sowie den aktuellen Stand in Bezug auf deren Identifizierung mittels NGS. Wir betrachten insbesondere die folgenden Fragen: Bei welchen häufigen Krankheiten ist ein Beitrag seltener Varianten zu erwarten, wie können diese Varianten identifiziert werden, und welches Potenzial bieten seltene Varianten für das Verständnis biologischer Prozesse bzw. für die Translation in die klinische Praxis?
Collapse
Affiliation(s)
- Kerstin U. Ludwig
- Aff2 0000 0000 8786 803X grid.15090.3d Emmy-Noether-Gruppe „Kraniofaziale Genomik“, Institut für Humangenetik U ni ver si täts kli ni kum Bonn Venusberg-Campus 1, Gebäude 76 53127 Bonn Deutschland
| | - Franziska Degenhardt
- Aff1 0000 0000 8786 803X grid.15090.3d Institut für Humangenetik Universitätsklinikum Bonn Bonn Deutschland
| | - Markus M. Nöthen
- Aff1 0000 0000 8786 803X grid.15090.3d Institut für Humangenetik Universitätsklinikum Bonn Bonn Deutschland
| |
Collapse
|
199
|
Romain K, Eriksson A, Onyon R, Kumar M. The psychosis risk timeline: can we improve our preventive strategies? Part 1: early life. BJPSYCH ADVANCES 2019. [DOI: 10.1192/bja.2018.66] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
SUMMARYPsychosis is a complex presentation with a wide range of factors contributing to its development, biological and environmental. Psychosis is a feature present in a variety of psychiatric disorders. It is important for clinicians to keep up to date with evidence regarding current understanding of the reasons psychosis may occur. Furthermore, it is necessary to find clinical utility from this knowledge so that effective primary, secondary and tertiary preventative strategies can be considered. This article is the first of a three-part series that examines contemporary knowledge of risk factors for psychosis and presents an overview of current explanations. The articles focus on the psychosis risk timeline, which gives a structure within which to consider key aspects of risk likely to affect people at different stages of life. In this first article, early life is discussed. It covers elements that contribute in the prenatal and early childhood period and includes genetic, nutritional and infective risk factors.LEARNING OBJECTIVESAfter reading this article you will be able to:
•give an up-to-date overview of psychosis risk factors that can affect early life•describe some important genetic risk factors•understand more about the role of environmental factors such as nutrition and infection.DECLARATION OF INTERESTNone.
Collapse
|
200
|
Cameron D, Blake DJ, Bray NJ, Hill MJ. Transcriptional Changes following Cellular Knockdown of the Schizophrenia Risk Gene SETD1A Are Enriched for Common Variant Association with the Disorder. MOLECULAR NEUROPSYCHIATRY 2019; 5:109-114. [PMID: 31192223 DOI: 10.1159/000497181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 01/22/2019] [Indexed: 01/01/2023]
Abstract
Loss of function mutations in SETD1A are the first experiment-wide significant findings to emerge from exome sequencing studies of schizophrenia. Although SETD1A is known to encode a histone methyltransferase, the consequences of reduced S ETD1A activity on gene expression in neural cells have, to date, been unknown. To explore transcriptional changes through which genetic perturbation of SETD1A could confer risk for schizophrenia, we have performed genome-wide gene expression profiling of a commonly used human neuroblastoma cell line in which SETD1A expression has been experimentally reduced using RNA interference (RNAi). We identified 1,031 gene expression changes that were significant in two separate RNAi conditions compared with control, including effects on genes of known neurodevelopmental importance such as DCX and DLX5. Genes that were differentially expressed following SETD1A knockdown were enriched for annotation to metabolic pathways, peptidase regulator activity and integrin-mediated regulation of cell adhesion. Moreover, differentially expressed genes were enriched for common variant association with schizophrenia, suggesting a degree of molecular convergence between this rare schizophrenia risk factor and susceptibility variants for the disorder operating more generally.
Collapse
Affiliation(s)
- Darren Cameron
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Derek J Blake
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Nicholas J Bray
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Matthew J Hill
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University School of Medicine, Cardiff, United Kingdom.,UK Dementia Research Institute at Cardiff University, Cardiff, United Kingdom.,Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|