Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Gene expression elucidates functional impact of polygenic risk for schizophrenia

Abstract

Over 100 genetic loci harbor schizophrenia-associated variants, yet how these variants confer liability is uncertain. The CommonMind Consortium sequenced RNA from dorsolateral prefrontal cortex of people with schizophrenia (N = 258) and control subjects (N = 279), creating a resource of gene expression and its genetic regulation. Using this resource, 20% of schizophrenia loci have variants that could contribute to altered gene expression and liability. In five loci, only a single gene was involved: FURIN, TSNARE1, CNTN4, CLCN3 or SNAP91. Altering expression of FURIN, TSNARE1 or CNTN4 changed neurodevelopment in zebrafish; knockdown of FURIN in human neural progenitor cells yielded abnormal migration. Of 693 genes showing significant case-versus-control differential expression, their fold changes were ≤ 1.33, and an independent cohort yielded similar results. Gene co-expression implicates a network relevant for schizophrenia. Our findings show that schizophrenia is polygenic and highlight the utility of this resource for mechanistic interpretations of genetic liability for brain diseases.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Enrichment of cis-eQTL in regulatory and other genomic elements.
Figure 2: Overlap of GWAS for schizophrenia with eQTL in the DLPFC.
Figure 3: Neuroanatomical phenotypes upon suppression or overexpression of genes at SCZ risk loci.
Figure 4: Decreasing FURIN expression in human NPCs perturbs neural migration.
Figure 5: Differential expression between schizophrenia cases and controls in the DLPFC.
Figure 6: Co-expression network analysis in control DLPFC samples.
Figure 7: Power to detect differential expression.

Similar content being viewed by others

Accession codes

Accessions

Gene Expression Omnibus

References

  1. McGrath, J., Saha, S., Chant, D. & Welham, J. Schizophrenia: a concise overview of incidence, prevalence, and mortality. Epidemiol. Rev. 30, 67–76 (2008).

    PubMed  Google Scholar 

  2. Kirov, G. CNVs in neuropsychiatric disorders. Hum. Mol. Genet. 24, R45–R49 (2015).

    CAS  PubMed  Google Scholar 

  3. Schizophrenia Working Group of the Psychiatric Genomics Consortium. Biological insights from 108 schizophrenia-associated genetic loci. Nature 511, 421–427 (2014).

  4. Purcell, S.M. et al. Common polygenic variation contributes to risk of schizophrenia and bipolar disorder. Nature 460, 748–752 (2009).

    CAS  PubMed  Google Scholar 

  5. Purcell, S.M. et al. A polygenic burden of rare disruptive mutations in schizophrenia. Nature 506, 185–190 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Walsh, T. et al. Rare structural variants disrupt multiple genes in neurodevelopmental pathways in schizophrenia. Science 320, 539–543 (2008).

    CAS  PubMed  Google Scholar 

  7. Fromer, M. et al. De novo mutations in schizophrenia implicate synaptic networks. Nature 506, 179–184 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Horváth, S., Janka, Z. & Mirnics, K. Analyzing schizophrenia by DNA microarrays. Biol. Psychiatry 69, 157–162 (2011).

    PubMed  PubMed Central  Google Scholar 

  9. Mistry, M., Gillis, J. & Pavlidis, P. Meta-analysis of gene coexpression networks in the post-mortem prefrontal cortex of patients with schizophrenia and unaffected controls. BMC Neurosci. 14, 105 (2013).

    PubMed  PubMed Central  Google Scholar 

  10. Hitzemann, R. et al. Introduction to sequencing the brain transcriptome. Int. Rev. Neurobiol. 116, 1–19 (2014).

    PubMed  PubMed Central  Google Scholar 

  11. Abecasis, G.R. et al. An integrated map of genetic variation from 1,092 human genomes. Nature 491, 56–65 (2012).

    PubMed  Google Scholar 

  12. Veyrieras, J.B. et al. High-resolution mapping of expression-QTLs yields insight into human gene regulation. PLoS Genet. 4, e1000214 (2008).

    PubMed  PubMed Central  Google Scholar 

  13. Dunham, I. et al. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    CAS  Google Scholar 

  14. GTEx Consortium. Human genomics. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science 348, 648–660 (2015).

  15. Zhang, B. et al. Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer's disease. Cell 153, 707–720 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Colantuoni, C. et al. Temporal dynamics and genetic control of transcription in the human prefrontal cortex. Nature 478, 519–523 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Gibbs, J.R. et al. Abundant quantitative trait loci exist for DNA methylation and gene expression in human brain. PLoS Genet. 6, e1000952 (2010).

    PubMed  PubMed Central  Google Scholar 

  18. Ramasamy, A. et al. Genetic variability in the regulation of gene expression in ten regions of the human brain. Nat. Neurosci. 17, 1418–1428 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Kim, Y. et al. A meta-analysis of gene expression quantitative trait loci in brain. Transl. Psychiatry 4, e459 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Wright, F.A. et al. Heritability and genomics of gene expression in peripheral blood. Nat. Genet. 46, 430–437 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Roussos, P. et al. A role for noncoding variation in schizophrenia. Cell Rep. 9, 1417–1429 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Richards, A.L. et al. Schizophrenia susceptibility alleles are enriched for alleles that affect gene expression in adult human brain. Mol. Psychiatry 17, 193–201 (2012).

    CAS  PubMed  Google Scholar 

  23. Trynka, G. et al. Chromatin marks identify critical cell types for fine mapping complex trait variants. Nat. Genet. 45, 124–130 (2013).

    CAS  PubMed  Google Scholar 

  24. Bharadwaj, R. et al. Conserved higher-order chromatin regulates NMDA receptor gene expression and cognition. Neuron 84, 997–1008 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. He, X. et al. Sherlock: detecting gene-disease associations by matching patterns of expression QTL and GWAS. Am. J. Hum. Genet. 92, 667–680 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. De Jager, P.L. et al. Alzheimer's disease: early alterations in brain DNA methylation at ANK1, BIN1, RHBDF2 and other loci. Nat. Neurosci. 17, 1156–1163 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Guzman, R.E., Alekov, A.K., Filippov, M., Hegermann, J. & Fahlke, C. Involvement of ClC-3 chloride/proton exchangers in controlling glutamatergic synaptic strength in cultured hippocampal neurons. Front. Cell. Neurosci. 8, 143 (2014).

    PubMed  PubMed Central  Google Scholar 

  28. Shimoda, Y. & Watanabe, K. Contactins: emerging key roles in the development and function of the nervous system. Cell Adh Migr 3, 64–70 (2009).

    PubMed  PubMed Central  Google Scholar 

  29. Kaneko-Goto, T., Yoshihara, S., Miyazaki, H. & Yoshihara, Y. BIG-2 mediates olfactory axon convergence to target glomeruli. Neuron 57, 834–846 (2008).

    CAS  PubMed  Google Scholar 

  30. Glessner, J.T. et al. Autism genome-wide copy number variation reveals ubiquitin and neuronal genes. Nature 459, 569–573 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Yuan, Q. et al. Regulation of brain-derived neurotrophic factor exocytosis and gamma-aminobutyric acidergic interneuron synapse by the schizophrenia susceptibility gene Dysbindin-1. Biol. Psychiatry 80, 312–322 (2016).

    CAS  PubMed  Google Scholar 

  32. Sekar, A. et al. Schizophrenia risk from complex variation of complement component 4. Nature 530, 177–183 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Mishra-Gorur, K. et al. Mutations in KATNB1 cause complex cerebral malformations by disrupting asymmetrically dividing neural progenitors. Neuron 84, 1226–1239 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Golzio, C. et al. KCTD13 is a major driver of mirrored neuroanatomical phenotypes of the 16p11.2 copy number variant. Nature 485, 363–367 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Carvalho, C.M. et al. Dosage changes of a segment at 17p13.1 lead to intellectual disability and microcephaly as a result of complex genetic interaction of multiple genes. Am. J. Hum. Genet. 95, 565–578 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Borck, G. et al. BRF1 mutations alter RNA polymerase III-dependent transcription and cause neurodevelopmental anomalies. Genome Res. 25, 155–166 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Brennand, K.J. et al. Modelling schizophrenia using human induced pluripotent stem cells. Nature 473, 221–225 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Topol, A. et al. Altered WNT signaling in human induced pluripotent stem cell neural progenitor cells derived from four schizophrenia patients. Biol. Psychiatry 78, e29–e34 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Lee, I.S. et al. Characterization of molecular and cellular phenotypes associated with a heterozygous CNTNAP2 deletion using patient-derived hiPSC neural cells. NPJ Schizophr 1, 15019 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Delaloy, C. & Gao, F.B. A new role for microRNA-9 in human neural progenitor cells. Cell Cycle 9, 2913–2914 (2010).

    CAS  PubMed  Google Scholar 

  41. Xiao, R. & Boehnke, M. Quantifying and correcting for the winner's curse in genetic association studies. Genet. Epidemiol. 33, 453–462 (2009).

    PubMed  PubMed Central  Google Scholar 

  42. Dawson, L.A. & Porter, R.A. Progress in the development of neurokinin 3 modulators for the treatment of schizophrenia: molecule development and clinical progress. Future Med. Chem. 5, 1525–1546 (2013).

    CAS  PubMed  Google Scholar 

  43. de Souza Silva, M.A. et al. Neurokinin3 receptor as a target to predict and improve learning and memory in the aged organism. Proc. Natl. Acad. Sci. USA 110, 15097–15102 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Ouchi, Y. et al. Reduced adult hippocampal neurogenesis and working memory deficits in the Dgcr8-deficient mouse model of 22q11.2 deletion-associated schizophrenia can be rescued by IGF2. J. Neurosci. 33, 9408–9419 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Sakai, T. et al. Changes in density of calcium-binding-protein-immunoreactive GABAergic neurons in prefrontal cortex in schizophrenia and bipolar disorder. Neuropathology 28, 143–150 (2008).

    PubMed  Google Scholar 

  46. Carboni, L. & Domenici, E. Proteome effects of antipsychotic drugs: learning from preclinical models. Proteomics Clin. Appl. 10, 430–441 (2016).

    CAS  PubMed  Google Scholar 

  47. Voineagu, I. et al. Transcriptomic analysis of autistic brain reveals convergent molecular pathology. Nature 474, 380–384 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Torkamani, A., Dean, B., Schork, N.J. & Thomas, E.A. Coexpression network analysis of neural tissue reveals perturbations in developmental processes in schizophrenia. Genome Res. 20, 403–412 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Oldham, M.C. et al. Functional organization of the transcriptome in human brain. Nat. Neurosci. 11, 1271–1282 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Roussos, P., Katsel, P., Davis, K.L., Siever, L.J. & Haroutunian, V. A system-level transcriptomic analysis of schizophrenia using postmortem brain tissue samples. Arch. Gen. Psychiatry 69, 1205–1213 (2012).

    PubMed  Google Scholar 

  51. Wang, K., Li, M. & Hakonarson, H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 38, e164 (2010).

    PubMed  PubMed Central  Google Scholar 

  52. Powchik, P. et al. Postmortem studies in schizophrenia. Schizophr. Bull. 24, 325–341 (1998).

    CAS  PubMed  Google Scholar 

  53. Purohit, D.P. et al. Alzheimer disease and related neurodegenerative diseases in elderly patients with schizophrenia: a postmortem neuropathologic study of 100 cases. Arch. Gen. Psychiatry 55, 205–211 (1998).

    CAS  PubMed  Google Scholar 

  54. Kimoto, S., Bazmi, H.H. & Lewis, D.A. Lower expression of glutamic acid decarboxylase 67 in the prefrontal cortex in schizophrenia: contribution of altered regulation by Zif268. Am. J. Psychiatry 171, 969–978 (2014).

    PubMed  PubMed Central  Google Scholar 

  55. Glantz, L.A. & Lewis, D.A. Decreased dendritic spine density on prefrontal cortical pyramidal neurons in schizophrenia. Arch. Gen. Psychiatry 57, 65–73 (2000).

    CAS  PubMed  Google Scholar 

  56. Volk, D.W., Austin, M.C., Pierri, J.N., Sampson, A.R. & Lewis, D.A. Decreased glutamic acid decarboxylase67 messenger RNA expression in a subset of prefrontal cortical gamma-aminobutyric acid neurons in subjects with schizophrenia. Arch. Gen. Psychiatry 57, 237–245 (2000).

    CAS  PubMed  Google Scholar 

  57. Purcell, S. et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am. J. Hum. Genet. 81, 559–575 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. O'Connell, J. et al. A general approach for haplotype phasing across the full spectrum of relatedness. PLoS Genet. 10, e1004234 (2014).

    PubMed  PubMed Central  Google Scholar 

  59. Howie, B., Fuchsberger, C., Stephens, M., Marchini, J. & Abecasis, G.R. Fast and accurate genotype imputation in genome-wide association studies through pre-phasing. Nat. Genet. 44, 955–959 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Lee, A.B., Luca, D., Klei, L., Devlin, B. & Roeder, K. Discovering genetic ancestry using spectral graph theory. Genet. Epidemiol. 34, 51–59 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Luca, D. et al. On the use of general control samples for genome-wide association studies: genetic matching highlights causal variants. Am. J. Hum. Genet. 82, 453–463 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Robinson, M.D., McCarthy, D.J. & Smyth, G.K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    CAS  PubMed  Google Scholar 

  63. Robinson, M.D. & Oshlack, A. A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol. 11, R25 (2010).

    PubMed  PubMed Central  Google Scholar 

  64. San Lucas, F.A., Wang, G., Scheet, P. & Peng, B. Integrated annotation and analysis of genetic variants from next-generation sequencing studies with variant tools. Bioinformatics 28, 421–422 (2012).

    CAS  PubMed  Google Scholar 

  65. Feng, H., Zhang, X. & Zhang, C. mRIN for direct assessment of genome-wide and gene-specific mRNA integrity from large-scale RNA-sequencing data. Nat. Commun. 6, 7816 (2015).

    CAS  PubMed  Google Scholar 

  66. DeLuca, D.S. et al. RNA-SeQC: RNA-seq metrics for quality control and process optimization. Bioinformatics 28, 1530–1532 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Katz, Y., Wang, E.T., Airoldi, E.M. & Burge, C.B. Analysis and design of RNA sequencing experiments for identifying isoform regulation. Nat. Methods 7, 1009–1015 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Law, C.W., Chen, Y., Shi, W. & Smyth, G.K. voom: Precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 15, R29 (2014).

    PubMed  PubMed Central  Google Scholar 

  69. Huang, T. & Cai, Y.D. An information-theoretic machine learning approach to expression QTL analysis. PLoS One 8, e67899 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Stegle, O., Parts, L., Piipari, M., Winn, J. & Durbin, R. Using probabilistic estimation of expression residuals (PEER) to obtain increased power and interpretability of gene expression analyses. Nat. Protoc. 7, 500–507 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Kundaje, A. et al. Integrative analysis of 111 reference human epigenomes. Nature 518, 317–330 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Auton, A. et al. A global reference for human genetic variation. Nature 526, 68–74 (2015).

    PubMed  Google Scholar 

  73. McCarthy, S.E. et al. Microduplications of 16p11.2 are associated with schizophrenia. Nat. Genet. 41, 1223–1227 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Topol, A., Tran, N.N. & Brennand, K.J. A guide to generating and using hiPSC derived NPCs for the study of neurological diseases. J. Vis. Exp. 96, e52495 (2015).

    Google Scholar 

  75. Topol, A. et al. Dysregulation of miRNA-9 in a Subset of Schizophrenia Patient-Derived Neural Progenitor Cells. Cell Rep. 15, 1024–1036 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Moffat, J. et al. A lentiviral RNAi library for human and mouse genes applied to an arrayed viral high-content screen. Cell 124, 1283–1298 (2006).

    CAS  PubMed  Google Scholar 

  77. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    CAS  PubMed  Google Scholar 

  78. Liao, Y., Smyth, G.K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    CAS  PubMed  Google Scholar 

  79. Cheng, M.C. et al. Chronic treatment with aripiprazole induces differential gene expression in the rat frontal cortex. Int. J. Neuropsychopharmacol. 11, 207–216 (2008).

    CAS  PubMed  Google Scholar 

  80. Orsetti, M., Di Brisco, F., Rinaldi, M., Dallorto, D. & Ghi, P. Some molecular effectors of antidepressant action of quetiapine revealed by DNA microarray in the frontal cortex of anhedonic rats. Pharmacogenet. Genomics 19, 600–612 (2009).

    CAS  PubMed  Google Scholar 

  81. Ikeda, M. et al. Identification of novel candidate genes for treatment response to risperidone and susceptibility for schizophrenia: integrated analysis among pharmacogenomics, mouse expression, and genetic case-control association approaches. Biol. Psychiatry 67, 263–269 (2010).

    CAS  PubMed  Google Scholar 

  82. Fatemi, S.H., Folsom, T.D., Reutiman, T.J., Novak, J. & Engel, R.H. Comparative gene expression study of the chronic exposure to clozapine and haloperidol in rat frontal cortex. Schizophr. Res. 134, 211–218 (2012).

    PubMed  Google Scholar 

  83. Rizig, M.A. et al. A gene expression and systems pathway analysis of the effects of clozapine compared to haloperidol in the mouse brain implicates susceptibility genes for schizophrenia. J. Psychopharmacol. 26, 1218–1230 (2012).

    CAS  PubMed  Google Scholar 

  84. Kondo, M.A. et al. Unique pharmacological actions of atypical neuroleptic quetiapine: possible role in cell cycle/fate control. Transl. Psychiatry 3, e243 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Santoro, M.L. et al. Effect of antipsychotic drugs on gene expression in the prefrontal cortex and nucleus accumbens in the spontaneously hypertensive rat (SHR). Schizophr. Res. 157, 163–168 (2014).

    PubMed  Google Scholar 

  86. Shi, W., Oshlack, A. & Smyth, G.K. Optimizing the noise versus bias trade-off for Illumina whole genome expression BeadChips. Nucleic Acids Res. 38, e204 (2010).

    PubMed  PubMed Central  Google Scholar 

  87. Johnson, W.E., Li, C. & Rabinovic, A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics 8, 118–127 (2007).

    PubMed  Google Scholar 

  88. Kirov, G. et al. The penetrance of copy number variations for schizophrenia and developmental delay. Biol. Psychiatry 75, 378–385 (2014).

    CAS  PubMed  Google Scholar 

  89. Girard, S.L. et al. Increased exonic de novo mutation rate in individuals with schizophrenia. Nat. Genet. 43, 860–863 (2011).

    CAS  PubMed  Google Scholar 

  90. Xu, B. et al. De novo gene mutations highlight patterns of genetic and neural complexity in schizophrenia. Nat. Genet. 44, 1365–1369 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Gulsuner, S. et al. Spatial and temporal mapping of de novo mutations in schizophrenia to a fetal prefrontal cortical network. Cell 154, 518–529 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. McCarthy, S.E. et al. De novo mutations in schizophrenia implicate chromatin remodeling and support a genetic overlap with autism and intellectual disability. Mol. Psychiatry 19, 652–658 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. De Rubeis, S. et al. Synaptic, transcriptional and chromatin genes disrupted in autism. Nature 515, 209–215 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Jiang, Y.H. et al. Detection of clinically relevant genetic variants in autism spectrum disorder by whole-genome sequencing. Am. J. Hum. Genet. 93, 249–263 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Iossifov, I. et al. The contribution of de novo coding mutations to autism spectrum disorder. Nature 515, 216–221 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. de Ligt, J. et al. Diagnostic exome sequencing in persons with severe intellectual disability. N. Engl. J. Med. 367, 1921–1929 (2012).

    CAS  PubMed  Google Scholar 

  97. Hamdan, F.F. et al. De novo mutations in moderate or severe intellectual disability. PLoS Genet. 10, e1004772 (2014).

    PubMed  PubMed Central  Google Scholar 

  98. Rauch, A. et al. Range of genetic mutations associated with severe non-syndromic sporadic intellectual disability: an exome sequencing study. Lancet 380, 1674–1682 (2012).

    CAS  PubMed  Google Scholar 

  99. EuroEPINOMICS-RES Consortium. De novo mutations in synaptic transmission genes including DNM1 cause epileptic encephalopathies. Am. J. Hum. Genet. 95, 360–370 (2014).

  100. Lee, P.H., O'Dushlaine, C., Thomas, B. & Purcell, S.M. INRICH: interval-based enrichment analysis for genome-wide association studies. Bioinformatics 28, 1797–1799 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Storey, J.D. & Tibshirani, R. Statistical significance for genomewide studies. Proc. Natl. Acad. Sci. USA 100, 9440–9445 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Tansey, K.E., Owen, M.J. & O'Donovan, M.C. Schizophrenia genetics: building the foundations of the future. Schizophr. Bull. 41, 15–19 (2015).

    PubMed  Google Scholar 

  103. Darnell, J.C. et al. FMRP stalls ribosomal translocation on mRNAs linked to synaptic function and autism. Cell 146, 247–261 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Ripke, S. et al. Genome-wide association analysis identifies 13 new risk loci for schizophrenia. Nat. Genet. 45, 1150–1159 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Kirov, G. et al. De novo CNV analysis implicates specific abnormalities of postsynaptic signalling complexes in the pathogenesis of schizophrenia. Mol. Psychiatry 17, 142–153 (2012).

    CAS  PubMed  Google Scholar 

  106. Ashburner, M. et al. Gene ontology: tool for the unification of biology. Nat. Genet. 25, 25–29 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Croft, D. et al. The Reactome pathway knowledgebase. Nucleic Acids Res. 42, D472–D477 (2014).

    CAS  PubMed  Google Scholar 

  108. Gray, K.A., Yates, B., Seal, R.L., Wright, M.W. & Bruford, E.A. Genenames.org: the HGNC resources in 2015. Nucleic Acids Res. 43, D1079–D1085 (2015).

    CAS  PubMed  Google Scholar 

  109. Goeman, J.J. & Bühlmann, P. Analyzing gene expression data in terms of gene sets: methodological issues. Bioinformatics 23, 980–987 (2007).

    CAS  PubMed  Google Scholar 

  110. Barbie, D.A. et al. Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1. Nature 462, 108–112 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Young, M.D., Wakefield, M.J., Smyth, G.K. & Oshlack, A. Gene ontology analysis for RNA-seq: accounting for selection bias. Genome Biol. 11, R14 (2010).

    PubMed  PubMed Central  Google Scholar 

  112. Hänzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7 (2013).

    PubMed  PubMed Central  Google Scholar 

  113. Tomfohr, J., Lu, J. & Kepler, T.B. Pathway level analysis of gene expression using singular value decomposition. BMC Bioinformatics 6, 225 (2005).

    PubMed  PubMed Central  Google Scholar 

  114. Lee, E., Chuang, H.Y., Kim, J.W., Ideker, T. & Lee, D. Inferring pathway activity toward precise disease classification. PLoS Comput. Biol. 4, e1000217 (2008).

    PubMed  PubMed Central  Google Scholar 

  115. Gentleman, R.C. et al. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 5, R80 (2004).

    PubMed  PubMed Central  Google Scholar 

  116. Brown, M.B. 400: A method for combining non-independent, one-sides tests of significance. Biometrics 31, 987–992 (1975).

    Google Scholar 

  117. Zhang, B. & Horvath, S. A general framework for weighted gene co-expression network analysis. Stat. Appl. Genet. Mol. Biol. 4, e17 (2005).

    Google Scholar 

  118. Ravasz, E., Somera, A.L., Mongru, D.A., Oltvai, Z.N. & Barabási, A.L. Hierarchical organization of modularity in metabolic networks. Science 297, 1551–1555 (2002).

    CAS  PubMed  Google Scholar 

  119. Langfelder, P. & Horvath, S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 9, 559 (2008).

    PubMed  PubMed Central  Google Scholar 

  120. Lambert, J.C. et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer's disease. Nat. Genet. 45, 1452–1458 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Okada, Y. et al. Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature 506, 376–381 (2014).

    CAS  PubMed  Google Scholar 

  122. Langfelder, P., Luo, R., Oldham, M.C. & Horvath, S. Is my network module preserved and reproducible? PLoS Comput. Biol. 7, e1001057 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Lein, E.S. et al. Genome-wide atlas of gene expression in the adult mouse brain. Nature 445, 168–176 (2007).

    CAS  PubMed  Google Scholar 

  124. Zhang, Y. et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J. Neurosci. 34, 11929–11947 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Bachoo, R.M. et al. Molecular diversity of astrocytes with implications for neurological disorders. Proc. Natl. Acad. Sci. USA 101, 8384–8389 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Foster, L.J. et al. A mammalian organelle map by protein correlation profiling. Cell 125, 187–199 (2006).

    CAS  PubMed  Google Scholar 

  127. Morciano, M. et al. Immunoisolation of two synaptic vesicle pools from synaptosomes: a proteomics analysis. J. Neurochem. 95, 1732–1745 (2005).

    CAS  PubMed  Google Scholar 

  128. Sugino, K. et al. Molecular taxonomy of major neuronal classes in the adult mouse forebrain. Nat. Neurosci. 9, 99–107 (2006).

    CAS  PubMed  Google Scholar 

  129. Winden, K.D. et al. The organization of the transcriptional network in specific neuronal classes. Mol. Syst. Biol. 5, 291 (2009).

    PubMed  PubMed Central  Google Scholar 

  130. Hawrylycz, M.J. et al. An anatomically comprehensive atlas of the adult human brain transcriptome. Nature 489, 391–399 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Oldham, M.C., Horvath, S. & Geschwind, D.H. Conservation and evolution of gene coexpression networks in human and chimpanzee brains. Proc. Natl. Acad. Sci. USA 103, 17973–17978 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Miller, J.A., Horvath, S. & Geschwind, D.H. Divergence of human and mouse brain transcriptome highlights Alzheimer disease pathways. Proc. Natl. Acad. Sci. USA 107, 12698–12703 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Chen, C. et al. Two gene co-expression modules differentiate psychotics and controls. Mol. Psychiatry 18, 1308–1314 (2013).

    CAS  PubMed  Google Scholar 

  134. de Jong, S. et al. A gene co-expression network in whole blood of schizophrenia patients is independent of antipsychotic-use and enriched for brain-expressed genes. PLoS One 7, e39498 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the patients and families who donated material for these studies. We thank T. Lehner for his early and inspirational ideas about this project, as well as organizational and intellectual support. We thank X. He for discussions regarding Sherlock, J. Scarpa for help running and interpreting WGCNA, L. Essioux for support in establishing and managing interactions with the Consortium, and A. Bertolino and A. Ghosh for continuous encouragement. Data and results were generated as part of the CommonMind Consortium supported by funding from Takeda Pharmaceuticals Company Limited, F. Hoffman-La Roche Ltd; grants R01MH093725-02S1 (J.D.B.), P50MH066392 (J.D.B.), R01MH097276 (P.S., E.E.S.), R01MH075916 (C.-G.H.), P50MH096891 C.-G.H., REG, P50MH084053-S1 (D.A.L.), R37MH057881 (B.D.), R37MH057881S1 (B.D.), R01MH085542-S1 (P.S.), U01MH096296-S2 (P.S.), HHSN271201300031C (V.H.), VA VISN3 MIRECC (V.H.), P50MH066392 (J.D.B.), NIMH Intramural program (B.K.L.), R01MH101454 (K.J.B.), R01MH109677 (P.R.), R01AG050986 (P.R.), VA Merit BX002395 (P.R.) and R01 AG036836 (P.D.H.); New York Stem Cell Foundation (K.J.B.); the Silvio O Conte Center grant P50MH094268 (N.K.); NARSAD (E.C.O.) and NARSAD Young Investigator (D.M.R., P.R., E.A.S.); the Stanley Medical Research Institute for Funding for Non-Human Primate Research; and NIMH grants R01MH074313 (S.E.H.); R01AG036836, U01AG046152 and R01AG017917 (D.A.B. and P.I.D.J.); R01AG046170 (E.S.S., B.Z., J.Z., and P.R.); and R01MH109706 (E.C.O.). Brain tissues for the study were obtained from the following brain bank collections: the Mount Sinai NIH Brain and Tissue Repository, the University of Pennsylvania Alzheimer's Disease Core Center, the University of Pittsburgh Brain Tissue Donation Program, the NIMH Human Brain Collection Core and Wake Forest University. CMC Leadership: P. Sklar and J.D. Buxbaum (Icahn School of Medicine at Mount Sinai), B. Devlin and D.A. Lewis (University of Pittsburgh), R.E. Gur and C.-G. Hahn (University of Pennsylvania), K. Hirai and H. Toyoshiba (Takeda Pharmaceuticals Company Limited), E. Domenici and L. Essioux (F. Hoffman-La Roche Ltd), L.M. Mangravite and M.A. Peters (Sage Bionetworks), and T. Lehner and B.K. Lipska (NIMH).

Author information

Authors and Affiliations

Authors

Contributions

P.R., J.S.J., K.T., R.E.G., C.-G.H., D.A.L., V.H., B.K.L. and J.D.B. contributed to sample collection. S.E.H. contributed monkey brain tissue and P.F.S. contributed mouse data. M.F., P.R., S.K.S., D.H.K., T.M.P., D.M.R., K.K.D., E.C.O., A.T., T.C., M.A.P., E.D., B.D. and P.S. contributed to the writing of this manuscript. M.F., P.R., S.K.S., H.R.S., D.M.R., K.K.D., M.C.M., J.M.J.D., A.C., S.M.P., L.A.S., L.M.M., H.T., D.A.L., M.A.P., J.D.B., E.E.S., K.H., K.J.B., N.K., B.D. and P.S. contributed to experimental and study design and planning analytical strategies. L.A.S., H.T., D.A.L., B.K.L., J.D.B., E.E.S., K.H., E.D., B.D. and P.S. contributed the funding of this work. M.F., P.R., S.K.S., J.S.J., D.H.K., T.M.P., D.M.R., H.R.S., L.L.K., R.K., D.P., Z.H.G., A.E.C., L.X., A.C., K.K.D., A.B., C.L., B.R., E.A.S., T.H., J.F.F., Y.-C.W., J.T.D., B.A.L., T.R., J.Z., B.Z., P.F.S., S.M.P., E.E.S., K.R., E.D., B.D. and P.S. contributed to data analyses. E.C.O., A.T., J.X., M.P., K.J.B. and N.K. contributed to the model system experiments. T.R., D.A.B., P.L.D.J. contributed the ROS/MAP data. A.C., L.A.S., L.M.M., H.T., R.E.G., C.-G.H., D.A.L., M.A.P., B.K.L., J.D.B., K.H., E.E.S., E.D., B.D. and P.S. contributed to the management and leadership of phase 1 of the CommonMind Consortium.

Corresponding author

Correspondence to Pamela Sklar.

Ethics declarations

Competing interests

E. Dominici was an employee of F. Hoffmann-La Roche for the first portion of the study and later served as a consultant to Roche in the area of genetic biomarkers. H. Toyoshiba and K. Hirai are employees of Takeda Pharmaceutical Company Limited and L.A. Shinobu is a former employee. D.A.L. currently receives investigator-initiated research support from Pfizer and from 2012 to 2014 served as a consultant in the areas of target identification and validation and new compound development to Autifony, Bristol-Myers Squibb, Concert Pharmaceuticals and Sunovion. M. Fromer was an employee of Mount Sinai until April 2016; he is now an employee of Google Verily.

Integrated supplementary information

Supplementary Figure 1 Experimental flow diagram and ancestry characterization.

(a) Samples were received from three brain banks and transferred to a single site for tissue processing, RNA sequencing, and DNA genotyping. Randomization of samples was performed at each of two steps: 1) prior to tissue processing and 2) again, prior to library preparation. (b) Distribution of genetic ancestry by brain bank, diagnosis, and nominal ancestry. (c) Ancestry dimension plots using assigned clusters (letters) and diagnosis (colors) to highlight additional features of the data. SCZ=blue, Control=red, Affective Disorders=orange. (d) Polygenic scoring profile for CMC. The x-axis gives the discovery p-value threshold (pT), and the y-axis the Nagelkerke’s case-control prediction R-squared.

Supplementary Figure 2 Analytic flow diagram and RNA sequencing metrics.

Top: Diagram of the mapping, QC and quantification of RNA sequencing, and basic QC results. Middle: Approach to data normalization and covariate adjustment. Bottom: Analytic strategy for differential expression, eQTLs, WGCNA with differential connectivity, and evaluation of genetic overlaps of results.

Supplementary Figure 3 qPCR validation of gene expression levels from RNA-seq.

(a) Comparisons of gene expression levels quantified using either qPCR (expression quantified as cycle number, normalized to that of beta actin, cyclophilin, and GAPDH) or voom-normalized log(CPM) levels from RNA-seq. Each point corresponds to one of N =114 individuals from the Pitt cohort: 57 SCZ cases (red) and 57 matched controls (blue). For each of the 13 genes validated, the corresponding gene symbol, Ensembl gene identifier, and Pearson correlation r between qPCR and RNA-seq are as shown. (b) Voom-normalized log(CPM) are presented by diagnosis and site for GAD1, PVALB, SLC32A1 and SST. Cases=pink, controls=green.

Supplementary Figure 4 Evaluation and selection of covariates and surrogate variables.

The steps to determine which covariates were used for gene expression adjustment (by linear modeling and regression) were: (a) Determine which “simple” (low degree of freedom) covariates add most to diagnosis as a predictor. (b) Test which batch effects (each with many levels) improve the model. (c) Cluster library batch effects to simplify them (reducing the number of degrees of freedom). (d) Re-cluster library batch effects after outlier sample removal. (e) Define the final library batch clusters (“clustered LIB”). (f) Evaluate the potential utility of quadratic terms. (g) Select the number of surrogate variables (SV) using SVA. (h) Test how many genes have variance better explained by addition of SV. (i) Summarize the model R2 explained (“model fit”) by either diagnosis alone, the base model including diagnosis (panel a), inclusion of the clustered library batch (b, e), adding in RIN2 (f), and adding in the SV (h); note the increasing R2 at each successive step of model selection, indicating that more of the gene expression variance is being captured by the model.

Supplementary Figure 5 Distribution of biological covariates.

(a) CMC and (b) HBCC

Supplementary Figure 6 Genome-wide overlap of GWAS for schizophrenia with eQTL in the DLPFC.

(a) Genome-wide Manhattan plot of Sherlock GWAS-eQTL overlap significance for all 12,367 genes (with one or more eQTL) tested for potential mediation of common variant risk of schizophrenia. Blue and red colors mark genes located on alternating chromosomes, and the dashed line denotes genome-wide Bonferroni significance (Pcorrected ≤ 0.05). (b) eQTL association profiles across two loci on chromosome 3 (highlighting CNTN4) and chromosome 8 (highlighting TSNARE1). Plot details are as in Fig. 2a.

Supplementary Figure 7 Summary of eQTL evidence pointing to single genes in GWAS loci.

(a) List of 10 genes (including 3 isoforms) ranked significantly by Sherlock for having gene and/or isoform eQTL profiles matching SCZ association, where no other gene (or gene isoform) in the corresponding GWAS locus is significant. For each gene and/or isoform, shown are the predicted direction of effect for the corresponding GWAS risk allele (predicted up-regulation in red and down-regulation in green), and whether the eQTL-to-GWAS correlation passes a manual visual assessment (see corresponding plots in panel B). (b) For each of the 13 genes or isoforms listed in A, the correlation between the significance (-log10(P)) of the eQTL associations at each eSNP (eQTL significance is to the right of the vertical dotted line) and the significance of association of that SNP with SCZ (genome-wide significance for GWAS is above the horizontal dotted line) is assessed. Each plot notes the number of total SNPs depicted (those with eQTL P < 0.1), the number of significantly associated eSNPs (P < 10-3 as input to Sherlock, or at FDR ≤ 5%), and Spearman’s rho (and p-value) for the correlation between the significant eQTL SNPs and their associations with SCZ. Plots are grouped into the 7 high quality single-gene GWAS-eQTL overlap results, the SNX19 gene that is high quality only at the isoform level, and two genes ranked highly by Sherlock but not passing this manual evaluation of correlation between disease and expression association signals.

Supplementary Figure 8 Validation of furin targeting.

(a) Schematic of genomic region on zebrafish furin_a gene targeted by a splice-blocking morpholino (MO) on the exon-intron region of exon 7. Position of RT-PCR primers for MO validation are also indicated. (b) RT-PCR assessment of furin_a MO efficiency. Only the MO-injected sample showed an amplicon containing the inclusion of intron 7 as a result of deleterious splicing caused by the MO. (c) Sanger sequencing confirming the inclusion of intron 7. (d) Schematic depicting the inclusion of intron 7 as a result of targeting furin_a. (e) Quantification of head size and proliferation defects caused by suppression of furin_a that can be rescued by co-expression of human FURIN mRNA. Error bars are s.e., * P < 0.05, ** P < 0.005, *** P < 0.0005.

Supplementary Figure 9 Correlation of differential expression between CMC and two meta-analytic studies.

Estimated differential expression for 23 genes with significant differential expression for CommonMind, after covariate adjustment, and for at least one of two meta-analytic studies. (a) CMC results fit with covariates as described in manuscript; (b) CMC results fit with only diagnosis as the covariate. Note that for the diagnosis-only model, none of these genes would be significant for CMC.

Supplementary Figure 10 In situ hybridization

Images are from the Allen Human Brain Atlas for selected genes among significant differentially expressed genes in CMC. Images are taken from the ISH data from the Neurotransmitter Study (176 genes across cortical regions and 88 genes across subcortical regions in 4 control cases). Images are taken from Frontal Cortex, Structure Name: middle frontal gyrus, right, Specimen H0351.1016 (White or Caucasian, Age 55 yrs, Male). Images suggest neuron specific expression for CALB1 (5.241) and GABRA5 (5.925); glial cell for ALDH1A1 (6.038) and SLC6A13 (1.512); endothelial cell expression for SLC38A5 (2.356); mixed expression for GABRB3 (7.795), predominantly neuronal; for GRIN3A (4.783) and CHRN2 (1.316), predominantly glial. The top right figure is a representative Nissl staining image of the same area. In brackets the average expression (pre-adjusted logCPM) determined by RNASeq in the CMC cohort is reported.

Supplementary Figure 11 Differential isoform expression.

(a) The number of isoforms per gene for all isoforms analyzed after filtering. (b) Analysis of differential expression of gene isoforms between SCZ and controls resulted in some consistency with results at the gene level, but also yielded additional genes that were not found to be differential when considered without isoform resolution. (c) Overlap between the genes differential at FDR ≤ 5% with the genes with isoforms differential at FDR ≤ 5%. (d) Prototypical examples for 4 genes with differential isoform expression (selected because each is found in the M2c co-expression module enriched for differential expression, see Fig. 6a). The mean log2 fold change, 95% confidence interval, and FDR value is plotted for each isoform. The top two genes (TBC1D15 and MSL1) each have at least one isoform differentially down-regulated in SCZ cases, but in aggregate (across all isoforms), the gene is not differentially expressed; for MSL1, this is likely due to the presence of 2 differential isoforms moving in opposite directions. On the other hand, the bottom two genes (N4BP2 and AFF2) are ranked as differential at the gene level, which seems to be mostly resulting from a single significant isoform in each instance.

Supplementary Figure 12 Cell type proportions.

Estimated cell fractions from human cortical tissue using (a) the program CIBERSORT to deconvolve the mixture based on 11,992 genes from the Zhang matrix of mouse cell-specific expression, (b) CIBERSORT to deconvolve the mixture based on 415 human cell-specific markers (markers estimated from the data), and (c) the CellMix package, lsfit option, using 11,992 genes from the Zhang matrix.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–12, Supplementary Information, and Supplementary Tables 1–3 (PDF 3496 kb)

Supplementary Methods Checklist (PDF 408 kb)

Supplementary Data File 1

Published post-mortem genome-wide differential gene expression studies in schizophrenia (XLSX 18 kb)

Supplementary Data File 2

Summary of regions with genes ranked highly by Sherlock; Genes with max-eQTL ranked as credible by PGC SCZ2 GWAS data; Isoforms with max-eQTL ranked as credible by PGC SCZ2 GWAS data (XLSX 31 kb)

Supplementary Data File 3

Differentially expressed genes; Differentially expressed isoforms; Comparison of CMC and HBCC (XLSX 1035 kb)

Supplementary Data File 4

Enrichment of differential expression in hypothesis-driven and hypothesis-free gene sets (XLSX 163 kb)

Supplementary Data File 5

Module assignments and connectivity values in the control network; Module assignments and connectivity values in the schizophrenia network (XLSX 2718 kb)

Supplementary Data File 6

Overlap of differentially expressed genes with modules in the control network; Overlap of differentially expressed genes with modules in the case network (XLSX 51 kb)

Supplementary Data File 7

Geneset enrichment for modules in the control network; Geneset enrichment for modules in the schizophrenia network (XLSX 157 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fromer, M., Roussos, P., Sieberts, S. et al. Gene expression elucidates functional impact of polygenic risk for schizophrenia. Nat Neurosci 19, 1442–1453 (2016). https://doi.org/10.1038/nn.4399

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4399

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research