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.

Gene expression imputation across multiple brain regions provides insights into schizophrenia risk

A Publisher Correction to this article was published on 13 May 2019

This article has been updated

Abstract

Transcriptomic imputation approaches combine eQTL reference panels with large-scale genotype data in order to test associations between disease and gene expression. These genic associations could elucidate signals in complex genome-wide association study (GWAS) loci and may disentangle the role of different tissues in disease development. We used the largest eQTL reference panel for the dorso-lateral prefrontal cortex (DLPFC) to create a set of gene expression predictors and demonstrate their utility. We applied DLPFC and 12 GTEx-brain predictors to 40,299 schizophrenia cases and 65,264 matched controls for a large transcriptomic imputation study of schizophrenia. We identified 413 genic associations across 13 brain regions. Stepwise conditioning identified 67 non-MHC genes, of which 14 did not fall within previous GWAS loci. We identified 36 significantly enriched pathways, including hexosaminidase-A deficiency, and multiple porphyric disorder pathways. We investigated developmental expression patterns among the 67 non-MHC genes and identified specific groups of pre- and postnatal expression.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Replication of DLPFC prediction models in independent data.
Fig. 2: SCZ association results.
Fig. 3: SCZ-associated genes are coexpressed throughout development and across brain regions.
Fig. 4: Gene expression patterns for SCZ-associated genes cluster into four groups, relating to distinct spatiotemporal expression.

Data availability

Our CMC-derived DLPFC prediction models are publicly available at https://github.com/laurahuckins/CMC_DLPFC_prediXcan.

Change history

  • 13 May 2019

    In the HTML version of the article originally published, the author group ‘The Schizophrenia Working Group of the Psychiatric Genomics Consortium’ was displayed incorrectly. The error has been corrected in the HTML version of the article.

References

  1. Nicolae, D. L. et al. Trait-associated SNPs are more likely to be eQTLs: annotation to enhance discovery from GWAS. PLoS Genet. 6, e1000888 (2010).

    PubMed  PubMed Central  Google Scholar 

  2. Dobbyn, A. et al. Co-localization of conditional eQTL and GWAS signatures in schizophrenia. Preprint at https://www.biorxiv.org/content/10.1101/129429v2 (2017).

  3. Gilad, Y., Rifkin, S. A. & Pritchard, J. K. Revealing the architecture of gene regulation: the promise of eQTL studies. Trends Genet. 24, 408–415 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Cookson, W., Liang, L., Abecasis, G., Moffatt, M. & Lathrop, M. Mapping complex disease traits with global gene expression. Nat. Rev. Genet. 10, 184–194 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Albert, F. W. & Kruglyak, L. The role of regulatory variation in complex traits and disease. Nat. Rev. Genet. 16, 197–212 (2015).

    CAS  PubMed  Google Scholar 

  6. Moffatt, M. F. et al. Genetic variants regulating ORMDL3 expression contribute to the risk of childhood asthma. Nature 448, 470–473 (2007).

    CAS  PubMed  Google Scholar 

  7. Speliotes, E. K. et al. Association analyses of 249,796 individuals reveal 18 new loci associated with body mass index. Nat. Genet. 42, 937–948 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Dubois, P. C. A. et al. Multiple common variants for celiac disease influencing immune gene expression. Nat. Genet. 42, 295–302 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Libioulle, C. et al. Novel Crohn disease locus identified by genome-wide association maps to a gene desert on 5p13.1 and modulates expression of PTGER4. PLoS Genet. 3, e58 (2007).

    PubMed  PubMed Central  Google Scholar 

  10. Giambartolomei, C. et al. Bayesian test for colocalisation between pairs of genetic association studies using summary statistics. PLoS Genet. 10, e1004383 (2014).

    PubMed  PubMed Central  Google Scholar 

  11. Boocock, J., Giambartolomei, C. & Stahl, E. A. COLOC2 (2016).

  12. Zhu, Z. et al. Integration of summary data from GWAS and eQTL studies predicts complex trait gene targets. Nat. Genet. 48, 481–487 (2016).

    CAS  PubMed  Google Scholar 

  13. Pickrell, J. K. et al. Detection and interpretation of shared genetic influences on 42 human traits. Nat. Genet. 48, 709–717 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Fromer, M. et al. Gene expression elucidates functional impact of polygenic risk for schizophrenia. Nat. Neurosci. 19, 1442–1453 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Gamazon, E. R. et al. A gene-based association method for mapping traits using reference transcriptome data. Nat. Genet. 47, 1091–1098 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Gusev, A. et al. Integrative approaches for large-scale transcriptome-wide association studies. Nat. Genet. 48, 245–252 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Geschwind, D. H. & Flint, J. Genetics and genomics of psychiatric disease. Science 349, 1489–94 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Leek, J. T., Johnson, W. E., Parker, H. S., Jaffe, A. E. & Storey, J. D. The sva package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics 28, 882–883 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Bennett, D. A., Schneider, J. A., Arvanitakis, Z. & Wilson, R. S. Overview and findings from the religious orders study. Curr. Alzheimer Res. 9, 628–645 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Bennett, D. A., Schneider, J. A., Buchman, A. S., Barnes, L. L. & Wilson, R. S. Overview and findings from the rush memory and aging project. Curr. Alzheimer Res. 9, 646–663 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Mele, M. et al. The human transcriptome across tissues and individuals. Science 348, 660–665 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Ripke, S. et al. Biological insights from 108 schizophrenia-associated genetic loci. Nature 511, 421–427 (2014).

    CAS  PubMed Central  Google Scholar 

  23. Dobbyn, A. et al. Landscape of conditional eQTL in dorsolateral prefrontal cortex and Co-localization with schizophrenia GWAS. Am. J. Hum. Genet. https://doi.org/10.1016/j.ajhg.2018.04.011(2018).

  24. Barbeira, A. N. et al. Exploring the phenotypic consequences of tissue specific gene expression variation inferred from GWAS summary statistics. Nat. Commun. 9, 1825 (2018).

    PubMed  PubMed Central  Google Scholar 

  25. Benjamin, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. Series B Stat. Methodol. 57, 289–300 (1995).

  26. 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 

  27. Fromer, M. et al. Gene expression elucidates functional impact of polygenic risk for schizophrenia. Nat. Neurosci. 19, 1442–1453 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Pardiñas, A. F. et al. Common schizophrenia alleles are enriched in mutation-intolerant genes and in regions under strong background selection. Nat. Genet. 50, 381–389 (2018).

    PubMed  PubMed Central  Google Scholar 

  29. Sanders, S. J. First glimpses of the neurobiology of autism spectrum disorder. Curr. Opin. Genet. Dev. 33, 80–92 (2015).

    CAS  PubMed  Google Scholar 

  30. Monkol, Lek. et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature 536, 285–291 (2016).

    Google Scholar 

  31. Malhotra, D. et al. High frequencies of de novo CNVs in bipolar disorder and schizophrenia. Neuron 72, 951–963 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Bautista, O., Vázquez-Caubet, J. C., Zhivago, E. A. & Dolores Sáiz, M. From metabolism to psychiatric symptoms: psychosis as a manifestation of acute intermittent porphyria. J. Neuropsychiatry Clin. Neurosci. 26, E30 (2014).

    PubMed  Google Scholar 

  33. Zimmermann, M., Bonaccurso, C., Valerius, C. & Hamann, G. F. Acute intermittent porphyria. A clinical chameleon: case study of a 40-year-old female patient. Nervenarzt 77, 1501–1505 (2006).

    CAS  PubMed  Google Scholar 

  34. Ventura, P. et al. A challenging diagnosis for potential fatal diseases: recommendations for diagnosing acute porphyrias. Eur. J. Intern. Med. 25, 497–505 (2014).

    PubMed  Google Scholar 

  35. Pischik, E. & Kauppinen, R. An update of clinical management of acute intermittent porphyria. Appl. Clin. Genet. 8, 201–214 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Kumar, B. Acute intermittent porphyria presenting solely with psychosis: a case report and discussion. Psychosomatics 53, 494–498 (2012).

    PubMed  Google Scholar 

  37. Bonnot, O. et al. Diagnostic and treatment implications of psychosis secondary to treatable metabolic disorders in adults: a systematic review. Orphanet J. Rare Dis. 9, 65 (2014).

    PubMed  PubMed Central  Google Scholar 

  38. Kaback, M. M. & Desnick, R. J. Hexosaminidase A Deficiency: GeneReviews (University of Washington, Seattle, 1993).

  39. Osama, S. Late onset Tay-Sachs disease presenting as a brief psychotic disorder with catatonia: a case report and review of literature. Jefferson J. Psych. 15, 4 (2000).

    Google Scholar 

  40. Skaper, S. D. in Brain Protection in Schizophrenia, Mood and Cognitive Disorders (ed. Ritsner, M. S.) 135–165 (Springer Science & Business Media, 2010).

  41. Castellano, E. et al. RAS signalling through PI3-Kinase controls cell migration via modulation of Reelin expression. Nat. Commun. 7, 11245 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Gururajan, A. & Buuse, M. van den. Is the mTOR-signalling cascade disrupted in Schizophrenia? J. Neurochem. 129, 377–387 (2014).

    CAS  PubMed  Google Scholar 

  43. Ritsner, M. S. Brain Protection in Schizophrenia, Mood and Cognitive Disorders (Springer Science & Business Media, 2010).

  44. Enriquez-Barreto, L. & Morales, M. The PI3K signaling pathway as a pharmacological target in Autism related disorders and Schizophrenia. Mol. Cell. Ther. 4, 2 (2016).

    PubMed  PubMed Central  Google Scholar 

  45. Glessner, J. T. et al. Strong synaptic transmission impact by copy number variations in schizophrenia. Proc. Natl Acad. Sci. USA 107, 10584–10589 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Network and Pathway Analysis Subgroup of Psychiatric Genomics Consortium. Psychiatric genome-wide association study analyses implicate neuronal, immune and histone pathways. Nat. Neurosci. 18, 199–209 (2015).

    Google Scholar 

  47. Bauman, A. L. et al. Cocaine and antidepressant-sensitive biogenic amine transporters exist in regulated complexes with protein phosphatase 2A. J. Neurosci. 20, 7571–7578 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Ayadi, A. et al. Mouse large-scale phenotyping initiatives: overview of the European Mouse Disease Clinic (EUMODIC) and of the Wellcome Trust Sanger Institute Mouse Genetics Project. Mamm. Genome 23, 600–610 (2012).

    PubMed  PubMed Central  Google Scholar 

  49. Keane, T. M. et al. Mouse genomic variation and its effect on phenotypes and gene regulation. Nature 477, 289–294 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Howe, D. G. et al. ZFIN, the zebrafish model organism database: increased support for mutants and transgenics. Nucleic Acids Res. 41, D854–D860 (2013).

    CAS  PubMed  Google Scholar 

  51. Smith, C. L., Blake, J. A., Kadin, J. A., Richardson, J. E. & Bult, C. J. Mouse genome database (MGD)-2018: knowledgebase for the laboratory mouse. Nucleic Acids Res. 46, D836–D842 (2018).

    CAS  PubMed  Google Scholar 

  52. Miller, J. A. et al. Transcriptional landscape of the prenatal human brain. Nature 508, 199–206 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Watts, D. J. & Strogatz, S. H. Collective dynamics of ‘small-world’ networks. Nature 393, 440–442 (1998).

    CAS  PubMed  Google Scholar 

  54. Nguyen, H. T. et al. Integrated Bayesian analysis of rare exonic variants to identify risk genes for schizophrenia and neurodevelopmental disorders. Genome Med. 9, 114 (2017).

    PubMed  PubMed Central  Google Scholar 

  55. Mancuso, N. et al. Integrating gene expression with summary association statistics to identify genes associated with 30 complex traits. Am. J. Hum. Genet. 100, 473–487 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Gottlieb, A., Daneshjou, R., DeGorter, M., Montgomery, S. & Altman, R. Population-specific imputation of gene expression improves prediction of pharmacogenomic traits for African Americans. Preprint at https://www.biorxiv.org/content/10.1101/115451v1 (2017).

  57. Need, A. & Goldstein, D. B. Next generation disparities in human genomics: concerns and remedies. Trends Genet 25, 489–494 (2009).

    CAS  PubMed  Google Scholar 

  58. Popejoy, A. & Fullerton, S. Genomics is failing on diversity. Nature 538, 161–164 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Pardiñas, A. F. et al. Common schizophrenia alleles are enriched in mutation-intolerant genes and maintained by background selection. Preprint at https://www.biorxiv.org/content/10.1101/068593v1 (2016).

  60. Browning, R. in The Poems of Robert Browning (eds Porter, C. & Clarke, H. A.) 257–271 (Thomas Y. Cromwell and Company, 1896).

  61. Loftus, L. S. & Arnold, W. N. Vincent van Gogh’s illness: acute intermittent porphyria? BMJ 303, 1589–1591 (1991).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Strik, W. K. The psychiatric illness of Vincent van Gogh. Nervenarzt 68, 401–409 (1997).

    CAS  PubMed  Google Scholar 

  63. Arnold, W. N. The illness of Vincent van Gogh. J. Hist. Neurosci. 13, 22–43 (2004).

    PubMed  Google Scholar 

  64. Hughes, J. R. A reappraisal of the possible seizures of Vincent van Gogh. Epilepsy Behav. 6, 504–510 (2005).

    PubMed  Google Scholar 

  65. Bhattacharyya, K. B. & Rai, S. The neuropsychiatric ailment of Vincent van Gogh. Ann. Indian Acad. Neurol. 18, 6–9 (2014).

    Google Scholar 

  66. Correa, R. Vincent van Gogh: A pathographic analysis. Med. Hypotheses 82, 141–144 (2014).

    CAS  PubMed  Google Scholar 

  67. Peters, T. J. & Beveridge, A. The madness of King George III: a psychiatric re-assessment. Hist. Psychiatry 21, 20–37 (2010).

    PubMed  Google Scholar 

  68. Szatkiewicz, J. P. et al. Copy number variation in schizophrenia in Sweden. Mol. Psychiatry 19, 762–773 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Cross-Disorder Group of the Psychiatric Genomics Consortium. Identification of risk loci with shared effects on five major psychiatric disorders: a genome-wide analysis. Lancet 381, 1371–1379 (2013).

  71. Keefe, R. S. E. & Fenton, W. S. How should DSM-V criteria for schizophrenia include cognitive impairment? Schizophr. Bull. 33, 912–920 (2007).

    PubMed  PubMed Central  Google Scholar 

  72. Reichenberg, A. et al. Static and dynamic cognitive deficits in childhood preceding adult schizophrenia: a 30-year study. Am. J. Psychiatry 167, 160–169 (2010).

    PubMed  PubMed Central  Google Scholar 

  73. Gold, J. M. Cognitive deficits as treatment targets in schizophrenia. Schizophr. Res. 72, 21–28 (2004).

    PubMed  Google Scholar 

  74. Cannon, M. et al. Evidence for early-childhood, pan-developmental impairment specific to schizophreniform disorder. Arch. Gen. Psychiatry 59, 449 (2002).

    PubMed  Google Scholar 

  75. Parikshak, N. N., Gandal, M. J., Geschwind, D. H. & Angeles, L. Systems biology and gene networks in neurodevelopmental and neurodegenerative disorders. Nat. Rev. Genet. 16, 441–458 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Glass, D. et al. Gene expression changes with age in skin, adipose tissue, blood and brain. Genome Biol. 14, R75 (2013).

    PubMed  PubMed Central  Google Scholar 

  77. Colantuoni, C. et al. Temporal dynamics and genetic control of transcription in the human prefrontal cortex. Nature 478, 519–523 (2012).

    Google Scholar 

  78. Gusev, A. et al. Transcriptome-wide association study of schizophrenia and chromatin activity yields mechanistic disease insights. Preprint at https://www.biorxiv.org/content/10.1101/067355v1 (2016).

  79. Ardlie, K. G. et al. The genotype-tissue expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science 348, 648–660 (2015).

  80. Willer, C. J., Li, Y. & Abecasis, G. R. METAL: fast and efficient meta-analysis of genomewide association scans. Bioinformatics 26, 2190–2191 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. COCHRAN, W. G. THE comparison of percentages in matched samplES. Biometrika 37, 256–266 (1950).

    CAS  PubMed  Google Scholar 

  82. Yang, J., Lee, S. H., Goddard, M. E. & Visscher, P. M. GCTA: a tool for genome-wide complex trait analysis. Am. J. Hum. Genet. 88, 76–82 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. de Leeuw, C. A., Mooij, J. M., Heskes, T. & Posthuma, D. MAGMA: generalized gene-set analysis of GWAS data. PLoS Comput. Biol. 11, e1004219 (2015).

    PubMed  PubMed Central  Google Scholar 

  84. 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 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  86. The Gene Ontology Consortium. Gene ontology consortium: going forward. Nucleic Acids Res. 43, D1049–D1056 (2014).

    PubMed Central  Google Scholar 

  87. Kanehisa, M. & Goto, S. KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28, 27–30 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  89. Thomas, P. D. et al. PANTHER: a library of protein families and subfamilies indexed by function. Genome Res. 13, 2129–2141 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Mi, H., Muruganujan, A. & Thomas, P. D. PANTHER in 2013: modeling the evolution of gene function, and other gene attributes, in the context of phylogenetic trees. Nucleic Acids Res. 41, D377–D386 (2013).

    CAS  PubMed  Google Scholar 

  91. Lin, G. N. et al. Spatiotemporal 16p11.2 Protein network implicates cortical late mid-fetal brain development and KCTD13- Cul3-RhoA pathway in psychiatric diseases. Neuron 85, 742–754 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Bahl, E., Koomar, T. & Michaelson, J. J. cerebroViz: An R package for anatomical visualization of spatiotemporal brain data. Bioinformatics 33, btw726 (2016).

    Google Scholar 

  93. van der Weyden, L., White, J. K., Adams, D. J. & Logan, D. W. The mouse genetics toolkit: revealing function and mechanism. Genome Biol. 12, 224 (2011).

    PubMed  PubMed Central  Google Scholar 

  94. Brown, S. D. M. & Moore, M. W. The international mouse phenotyping consortium: past and future perspectives on mouse phenotyping. Mamm. Genome 23, 632–640 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We dedicate this manuscript to the memory of Pamela Sklar, whose guidance and wisdom we miss daily. We strive to continue her legacy of thoughtful, innovative, and collaborative science. Data were generated as part of the CommonMind Consortium supported by funding from Takeda Pharmaceuticals Company Limited, F. Hoffman-La Roche Ltd and NIH grants R01MH085542, R01MH093725, P50MH066392, P50MH080405, R01MH097276, RO1-MH-075916, P50M096891, P50MH084053S1, R37MH057881 and R37MH057881S1, HHSN271201300031C, AG02219, AG05138 and MH06692.

Brain tissue for the study was 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 NeuroBioBank and Brain and Tissue Repositories and the NIMH Human Brain Collection Core. CMC Leadership: P. Sklar, J. Buxbaum (Icahn School of Medicine at Mount Sinai), B. Devlin, D. Lewis (University of Pittsburgh), R. Gur, C.-G. Hahn (University of Pennsylvania), K. Hirai, H. Toyoshiba (Takeda Pharmaceuticals Company Limited), E. Domenici, L. Essioux (F. Hoffman-La Roche Ltd), L. Mangravite, M. Peters (Sage Bionetworks), T. Lehner, B. Lipska (NIMH).

ROSMAP study data were provided by the Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago. Data collection was supported through funding by NIA grants P30AG10161, R01AG15819, R01AG17917, R01AG30146, R01AG36836, U01AG32984, U01AG46152, the Illinois Department of Public Health, and the Translational Genomics Research Institute.

The iPSYCH-GEMS team acknowledges funding from the Lundbeck Foundation (grant no. R102-A9118 and R155-2014-1724), the Stanley Medical Research Institute, an Advanced Grant from the European Research Council (project no. 294838), the Danish Strategic Research Council the Novo Nordisk Foundation for supporting the Danish National Biobank resource, and grants from Aarhus and Copenhagen Universities and University Hospitals, including support to the iSEQ Center, the GenomeDK HPC facility, and the CIRRAU Center.

The Genotype-Tissue Expression (GTEx) Project was supported by the Common Fund of the Office of the Director of the National Institutes of Health, and by NCI, NHGRI, NHLBI, NIDA, NIMH, and NINDS. The data used for the analyses described in this manuscript were obtained from the GTEx Portal on September 5, 2016. BrainSpan: Atlas of the Developing Human Brain (Internet). Funded by ARRA Awards 1RC2MH089921-01, 1RC2MH090047-01, and 1RC2MH089929-01.

H.K.I. was supported by R01 MH107666-01.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

L.M.H. designed the study and specific subanalyses, ran analyses, and wrote the manuscript. A.D. designed and ran analyses and contributed to the writing group. D.M.R. contributed to study and analytical design, and writing. G.H. contributed to analytical design and writing. W.W., H.T.N., and J.B. designed and ran specific analyses. A.F.P., V.M.R., T.D.A., K.G., M.F. all ran specific analyses. S.K.S. designed the study and analyses and contributed to the writing group. P.R. and R.K. designed the study and contributed data. E.D. designed the study, contributed data, and contributed to the writing group. E.R.G. designed specific analyses, and contributed to the writing group. S.P. designed the study. All three consortia (CMC, PGC-SCZ, iPSYCH-GEMS) contributed data. D.D., A.D.B., J.T.R.W., M.C.O’D., M.J.O. contributed data, advised on analyses, and contributed to the writing group. P. Sullivan advised on analyses and contributed to the writing group. B.D. designed the study, contributed data, advised on analyses, and contributed to the writing group. N.J.C. and H.K.I. designed the study, advised on analyses, and contributed to the writing group. P. Sklar and E.A.S. designed the study and specific analyses, ran analyses, and contributed to the writing group.

Corresponding author

Correspondence to Laura M. Huckins.

Ethics declarations

Competing interests

E.D. has received research support from Roche during 2016–2018. T.W. has acted as advisor and lecturer to H. Lundbeck A/S. All other authors declare no conflicts of interest.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Information

Supplementary Note and Supplementary Figures 1–12

Reporting Summary

Supplementary Table 1

Forward stepwise conditional analysis results.

Supplementary Table 2

MAGMA based pathway association results.

Supplementary Table 3

Mutant mouse lines lacking expression of SCZ-associated genes

Supplementary Table 4

Gene set membership of SCZ-associated genes, according to BRAINSPAN clusters

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Huckins, L.M., Dobbyn, A., Ruderfer, D.M. et al. Gene expression imputation across multiple brain regions provides insights into schizophrenia risk. Nat Genet 51, 659–674 (2019). https://doi.org/10.1038/s41588-019-0364-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-019-0364-4

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing