Increased burden of ultra-rare protein-altering variants among 4,877 individuals with schizophrenia

By analyzing the exomes of 12,332 unrelated Swedish individuals, including 4,877 individuals affected with schizophrenia, in ways informed by exome sequences from 45,376 other individuals, we identified 244,246 coding-sequence and splice-site ultra-rare variants (URVs) that were unique to individual Swedes. We found that gene-disruptive and putatively protein-damaging URVs (but not synonymous URVs) were more abundant among individuals with schizophrenia than among controls (P = 1.3 × 10−10). This elevation of protein-compromising URVs was several times larger than an analogously elevated rate for de novo mutations, suggesting that most rare-variant effects on schizophrenia risk are inherited. Among individuals with schizophrenia, the elevated frequency of protein-compromising URVs was concentrated in brain-expressed genes, particularly in neuronally expressed genes; most of this elevation arose from large sets of genes whose RNAs have been found to interact with synaptically localized proteins. Our results suggest that synaptic dysfunction may mediate a large fraction of strong, individually rare genetic influences on schizophrenia risk.

[1]  Stephan J Sanders,et al.  A framework for the interpretation of de novo mutation in human disease , 2014, Nature Genetics.

[2]  C. Sander,et al.  Predicting the functional impact of protein mutations: application to cancer genomics , 2011, Nucleic acids research.

[3]  Kenny Q. Ye,et al.  Strong Association of De Novo Copy Number Mutations with Autism , 2007, Science.

[4]  Peter Holmans,et al.  De novo CNVs in bipolar affective disorder and schizophrenia , 2014, Human molecular genetics.

[5]  Alan F. Scott,et al.  Online Mendelian Inheritance in Man (OMIM), a knowledgebase of human genes and genetic disorders , 2004, Nucleic Acids Res..

[6]  S. Grant,et al.  Characterization of the proteome, diseases and evolution of the human postsynaptic density , 2011, Nature Neuroscience.

[7]  T. Crow Is Psychosis a Disorder of XY Epigenetics? , 2015, EBioMedicine.

[8]  James Y. Zou Analysis of protein-coding genetic variation in 60,706 humans , 2015, Nature.

[9]  E. Boerwinkle,et al.  dbNSFP v2.0: A Database of Human Non‐synonymous SNVs and Their Functional Predictions and Annotations , 2013, Human mutation.

[10]  J. McGrath,et al.  Schizophrenia: a concise overview of incidence, prevalence, and mortality. , 2008, Epidemiologic reviews.

[11]  P. McGuffin,et al.  Fecundity of patients with schizophrenia, autism, bipolar disorder, depression, anorexia nervosa, or substance abuse vs their unaffected siblings. , 2013, JAMA psychiatry.

[12]  Pak Chung Sham,et al.  Genetic Power Calculator: design of linkage and association genetic mapping studies of complex traits , 2003, Bioinform..

[13]  J. Nielsen,et al.  Analysis of the Human Tissue-specific Expression by Genome-wide Integration of Transcriptomics and Antibody-based Proteomics* , 2013, Molecular & Cellular Proteomics.

[14]  E. Banks,et al.  De novo mutations in schizophrenia implicate synaptic networks , 2014, Nature.

[15]  B. V. van Bon,et al.  Diagnostic exome sequencing in persons with severe intellectual disability. , 2012, The New England journal of medicine.

[16]  Michael R. Johnson,et al.  De novo mutations in synaptic transmission genes including DNM1 cause epileptic encephalopathies. , 2014, American journal of human genetics.

[17]  Simon C. Potter,et al.  Genome-wide Association Analysis Identifies 14 New Risk Loci for Schizophrenia , 2013, Nature Genetics.

[18]  M. Owen,et al.  Exome arrays capture polygenic rare variant contributions to schizophrenia , 2016, Human molecular genetics.

[19]  J. Moeschler Genetic evaluation of intellectual disabilities. , 2008, Seminars in pediatric neurology.

[20]  Eric S. Lander,et al.  A polygenic burden of rare disruptive mutations in schizophrenia , 2014, Nature.

[21]  Tao Wang,et al.  SynaptomeDB: an ontology-based knowledgebase for synaptic genes , 2012, Bioinform..

[22]  Elvira Bramon,et al.  Disruption of the neurexin 1 gene is associated with schizophrenia. , 2009, Human molecular genetics.

[23]  Terrence J. Sejnowski,et al.  Epigenomic Signatures of Neuronal Diversity in the Mammalian Brain , 2015, Neuron.

[24]  Michael R. Johnson,et al.  De novo mutations in the classic epileptic encephalopathies , 2013, Nature.

[25]  M. Daly,et al.  Autism spectrum disorder severity reflects the average contribution of de novo and familial influences , 2014, Proceedings of the National Academy of Sciences.

[26]  M. Daly,et al.  Searching for missing heritability: Designing rare variant association studies , 2014, Proceedings of the National Academy of Sciences.

[27]  Martin S. Taylor,et al.  Disruption of two novel genes by a translocation co-segregating with schizophrenia. , 2000, Human molecular genetics.

[28]  Jana Marie Schwarz,et al.  MutationTaster evaluates disease-causing potential of sequence alterations , 2010, Nature Methods.

[29]  Pablo Cingolani,et al.  Using Drosophila melanogaster as a Model for Genotoxic Chemical Mutational Studies with a New Program, SnpSift , 2012, Front. Gene..

[30]  I. Ionita-Laza,et al.  Loss-of-Function Variants in Schizophrenia Risk and SETD1A as a Candidate Susceptibility Gene , 2014, Neuron.

[31]  T. Crow The XY Gene Hypothesis of Psychosis: Origins and Current Status , 2013, American journal of medical genetics. Part B, Neuropsychiatric genetics : the official publication of the International Society of Psychiatric Genetics.

[32]  Pablo Cingolani,et al.  © 2012 Landes Bioscience. Do not distribute. , 2022 .

[33]  Boris Yamrom,et al.  Rare De Novo and Transmitted Copy-Number Variation in Autistic Spectrum Disorders , 2011, Neuron.

[34]  S. Henikoff,et al.  Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm , 2009, Nature Protocols.

[35]  K. Nagata,et al.  Biochemical and morphological characterization of MAGI‐1 in neuronal tissue , 2012, Journal of neuroscience research.

[36]  John Wei,et al.  Copy number variant study of bipolar disorder in Canadian and UK populations implicates synaptic genes , 2014, American journal of medical genetics. Part B, Neuropsychiatric genetics : the official publication of the International Society of Psychiatric Genetics.

[37]  Xianjin Zhou,et al.  Over-expression of XIST, the Master Gene for X Chromosome Inactivation, in Females With Major Affective Disorders , 2015, EBioMedicine.

[38]  C. Chen,et al.  JARID1B is a histone H3 lysine 4 demethylase up-regulated in prostate cancer , 2007, Proceedings of the National Academy of Sciences.

[39]  Joshua C Randall,et al.  Prevalence, phenotype and architecture of developmental disorders caused by de novo mutation: The Deciphering Developmental Disorders Study , 2016, bioRxiv.

[40]  Iuliana Ionita-Laza,et al.  Sequence kernel association tests for the combined effect of rare and common variants. , 2013, American journal of human genetics.

[41]  J. MacCabe,et al.  A systematic review and meta‐analysis of the fertility of patients with schizophrenia and their unaffected relatives , 2011, Acta psychiatrica Scandinavica.

[42]  D. Pinto,et al.  Structural variation of chromosomes in autism spectrum disorder. , 2008, American journal of human genetics.

[43]  C. Spencer,et al.  A contribution of novel CNVs to schizophrenia from a genome-wide study of 41,321 subjects: CNV Analysis Group and the Schizophrenia Working Group of the Psychiatric Genomics Consortium , 2016, bioRxiv.

[44]  S Purcell,et al.  De novo CNV analysis implicates specific abnormalities of postsynaptic signalling complexes in the pathogenesis of schizophrenia , 2011, Molecular Psychiatry.

[45]  D. Licatalosi,et al.  FMRP Stalls Ribosomal Translocation on mRNAs Linked to Synaptic Function and Autism , 2011, Cell.

[46]  J. Nielsen,et al.  Analysis of the Human Tissue-specific Expression by Genome-wide Integration of Transcriptomics and Antibody-based Proteomics. , 2014, Molecular & cellular proteomics : MCP.

[47]  D. Horn,et al.  Range of genetic mutations associated with severe non-syndromic sporadic intellectual disability: an exome sequencing study , 2012, The Lancet.

[48]  Carolyn J. Brown,et al.  Analysis of expressed SNPs identifies variable extents of expression from the human inactive X chromosome , 2013, Genome Biology.

[49]  L. Siever,et al.  Spatial and Temporal Mapping of De Novo Mutations in Schizophrenia to a Fetal Prefrontal Cortical Network , 2013, Cell.

[50]  E. Boerwinkle,et al.  dbNSFP: A Lightweight Database of Human Nonsynonymous SNPs and Their Functional Predictions , 2011, Human mutation.

[51]  C. Spencer,et al.  Biological Insights From 108 Schizophrenia-Associated Genetic Loci , 2014, Nature.

[52]  J. Moeschler,et al.  Clinical Genetic Evaluation of the Child With Mental Retardation or Developmental Delays , 2006, Pediatrics.

[53]  P. Visscher,et al.  GCTA: a tool for genome-wide complex trait analysis. , 2011, American journal of human genetics.

[54]  Anjali J. Koppal,et al.  Supplementary data: Comprehensive modeling of microRNA targets predicts functional non-conserved and non-canonical sites , 2010 .

[55]  K. Mikoshiba,et al.  Ataxia and epileptic seizures in mice lacking type 1 inositol 1,4,5-trisphosphate receptor , 1996, Nature.

[56]  P. Visscher,et al.  Common polygenic variation contributes to risk of schizophrenia and bipolar disorder , 2009, Nature.

[57]  D. Geschwind,et al.  Cytoplasmic Rbfox1 Regulates the Expression of Synaptic and Autism-Related Genes , 2016, Neuron.

[58]  Boris Yamrom,et al.  The contribution of de novo coding mutations to autism spectrum disorder , 2014, Nature.

[59]  Y. Xing,et al.  A Transcriptome Database for Astrocytes, Neurons, and Oligodendrocytes: A New Resource for Understanding Brain Development and Function , 2008, The Journal of Neuroscience.

[60]  J. Gécz,et al.  The genetic landscape of intellectual disability arising from chromosome X. , 2009, Trends in genetics : TIG.

[61]  David Haussler,et al.  The UCSC Known Genes , 2006, Bioinform..

[62]  Deborah A Nickerson,et al.  De novo rates and selection of large copy number variation. , 2010, Genome research.

[63]  Carson C Chow,et al.  Second-generation PLINK: rising to the challenge of larger and richer datasets , 2014, GigaScience.

[64]  M. DePristo,et al.  A framework for variation discovery and genotyping using next-generation DNA sequencing data , 2011, Nature Genetics.

[65]  L. Vissers,et al.  Genome sequencing identifies major causes of severe intellectual disability , 2014, Nature.

[66]  Jacy L. Wagnon,et al.  CELF4 Regulates Translation and Local Abundance of a Vast Set of mRNAs, Including Genes Associated with Regulation of Synaptic Function , 2012, PLoS genetics.

[67]  Manuel A. R. Ferreira,et al.  PLINK: a tool set for whole-genome association and population-based linkage analyses. , 2007, American journal of human genetics.

[68]  S. Levy,et al.  De novo gene mutations highlight patterns of genetic and neural complexity in schizophrenia , 2012, Nature Genetics.

[69]  J. Stockman,et al.  Common genetic determinants of schizophrenia and bipolar disorder in Swedish families: a population-based study , 2010 .

[70]  J. Gécz,et al.  Mutations in the JARID1C gene, which is involved in transcriptional regulation and chromatin remodeling, cause X-linked mental retardation. , 2005, American journal of human genetics.

[71]  L. Maquat Nonsense-mediated mRNA decay: splicing, translation and mRNP dynamics , 2004, Nature Reviews Molecular Cell Biology.

[72]  Edouard Henrion,et al.  De Novo Mutations in Moderate or Severe Intellectual Disability , 2014, PLoS genetics.

[73]  P. Sullivan,et al.  Schizophrenia as a complex trait: evidence from a meta-analysis of twin studies. , 2003, Archives of general psychiatry.

[74]  Steven E. Hyman,et al.  Genetic research in autism spectrum disorders , 2015, Current opinion in pediatrics.

[75]  Adam J. Schwarz,et al.  CNVs conferring risk of autism or schizophrenia affect cognition in controls , 2013, Nature.

[76]  Murim Choi,et al.  De novo mutations in histone modifying genes in congenital heart disease , 2013, Nature.

[77]  M. DePristo,et al.  The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. , 2010, Genome research.

[78]  M C O'Donovan,et al.  Copy number variation in schizophrenia in Sweden , 2014, Molecular Psychiatry.

[79]  Gary D Bader,et al.  Functional impact of global rare copy number variation in autism spectrum disorders , 2010, Nature.

[80]  Kathryn Roeder,et al.  Multiple Recurrent De Novo CNVs, Including Duplications of the 7q11.23 Williams Syndrome Region, Are Strongly Associated with Autism , 2011, Neuron.

[81]  Mauricio O. Carneiro,et al.  From FastQ Data to High‐Confidence Variant Calls: The Genome Analysis Toolkit Best Practices Pipeline , 2013, Current protocols in bioinformatics.

[82]  S. Scherer,et al.  Detection of clinically relevant genetic variants in autism spectrum disorder by whole-genome sequencing. , 2013, American journal of human genetics.

[83]  T. N. Bhat,et al.  The Protein Data Bank , 2000, Nucleic Acids Res..

[84]  Tom R. Gaunt,et al.  Predicting the Functional, Molecular, and Phenotypic Consequences of Amino Acid Substitutions using Hidden Markov Models , 2012, Human mutation.

[85]  Thomas Bourgeron,et al.  Mapping autism risk loci using genetic linkage and chromosomal rearrangements , 2007, Nature Genetics.

[86]  P. Bork,et al.  A method and server for predicting damaging missense mutations , 2010, Nature Methods.

[87]  Kenny Q. Ye,et al.  An integrated map of genetic variation from 1,092 human genomes , 2012, Nature.

[88]  Stephen J. Guter,et al.  Convergence of Genes and Cellular Pathways Dysregulated in Autism Spectrum Disorders , 2014, American journal of human genetics.

[89]  J. Miller,et al.  Predicting the Functional Effect of Amino Acid Substitutions and Indels , 2012, PloS one.

[90]  Jakob Grove,et al.  Genetic risk for autism spectrum disorders and neuropsychiatric variation in the general population , 2015, Nature Genetics.

[91]  Joseph A. Gogos,et al.  Strong association of de novo copy number mutations with sporadic schizophrenia , 2008, Nature Genetics.

[92]  Giulio Genovese,et al.  Schizophrenia risk from complex variation of complement component 4 , 2016, Nature.

[93]  Anders D. Børglum,et al.  Genome-wide association study identifies five new schizophrenia loci , 2011, Nature Genetics.

[94]  Michael Wigler,et al.  Rare De Novo Variants Associated with Autism Implicate a Large Functional Network of Genes Involved in Formation and Function of Synapses , 2011, Neuron.

[95]  De novo mutations in epileptic encephalopathies , 2013 .

[96]  Yakir A Reshef,et al.  Partitioning heritability by functional annotation using genome-wide association summary statistics , 2015, Nature Genetics.

[97]  Michael Q. Zhang,et al.  HITS-CLIP and integrative modeling define the Rbfox splicing-regulatory network linked to brain development and autism. , 2014, Cell reports.

[98]  P. Visscher,et al.  Modeling Linkage Disequilibrium Increases Accuracy of Polygenic Risk Scores , 2015, bioRxiv.

[99]  Evan T. Geller,et al.  Patterns and rates of exonic de novo mutations in autism spectrum disorders , 2012, Nature.

[100]  S. Gabriel,et al.  Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. , 2014, The New England journal of medicine.

[101]  Alan F. Scott,et al.  Online Mendelian Inheritance in Man (OMIM), a knowledgebase of human genes and genetic disorders , 2002, Nucleic Acids Res..

[102]  S. Lok,et al.  Increased exonic de novo mutation rate in individuals with schizophrenia , 2011, Nature Genetics.

[103]  R. Stevenson,et al.  Atlas of X-Linked Intellectual Disability Syndromes , 2012 .

[104]  Juliane Hoyer,et al.  Diagnostic yield of various genetic approaches in patients with unexplained developmental delay or mental retardation , 2006, American journal of medical genetics. Part A.

[105]  Seungtai Yoon,et al.  De novo Mutations in Schizophrenia Implicate Chromatin Remodeling and Support a Genetic Overlap with Autism and Intellectual Disability , 2014, Molecular Psychiatry.

[106]  Minna Männikkö,et al.  Rare loss-of-function variants in SETD1A are associated with schizophrenia and developmental disorders , 2016, Nature Neuroscience.

[107]  Epilepsy Phenome,et al.  De novo mutations in synaptic transmission genes including DNM1 cause epileptic encephalopathies. , 2014, American journal of human genetics.

[108]  Justin C. Fay,et al.  Identification of deleterious mutations within three human genomes. , 2009, Genome research.

[109]  Christopher S. Poultney,et al.  Synaptic, transcriptional, and chromatin genes disrupted in autism , 2014, Nature.

[110]  S. Kawamoto,et al.  Biochemical and morphological characterization of A2BP1 in neuronal tissue , 2013, Journal of neuroscience research.

[111]  Richard Durbin,et al.  Sequence analysis Fast and accurate short read alignment with Burrows – Wheeler transform , 2009 .

[112]  R. Handsaker,et al.  Genome-wide association study in a Swedish population yields support for greater CNV and MHC involvement in schizophrenia compared with bipolar disorder , 2012, Molecular Psychiatry.

[113]  J. Sebat,et al.  High Frequencies of De Novo CNVs in Bipolar Disorder and Schizophrenia , 2011, Neuron.

[114]  Kenny Q. Ye,et al.  De Novo Gene Disruptions in Children on the Autistic Spectrum , 2012, Neuron.

[115]  Joseph K. Pickrell,et al.  A Systematic Survey of Loss-of-Function Variants in Human Protein-Coding Genes , 2012, Science.