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.

  • Review Article
  • Published:

The road to precision psychiatry: translating genetics into disease mechanisms

Abstract

Hundreds of genetic loci increasing risk for neuropsychiatric disorders have recently been identified. This success, perhaps paradoxically, has posed challenges for therapeutic development, which are amplified by the highly polygenic and pleiotropic nature of these genetic contributions. Success requires understanding the biological impact of single genetic variants and predicting their effects within an individual. Comprehensive functional genomic annotation of risk loci provides a framework for interpretation of neurobiological impact, requiring experimental validation with in vivo or in vitro model systems. Systems-level, integrative pathway analyses are beginning to elucidate the additive, polygenic contributions of risk variants on specific cellular, molecular, developmental, or circuit-level processes. Although most neuropsychiatric disease modeling has focused on genes disrupted by rare, large-effect-size mutations, common smaller-effect-size variants may also provide solid therapeutic targets to inform precision medicine approaches. Here we enumerate the promise and challenges of a genomics-driven approach to uncovering neuropsychiatric disease mechanisms and facilitating therapeutic development.

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

Figure 1: Genetic and environmental contribution to liability for neuropsychiatric disease.
Figure 2: Neurobiological framework for interpretation of individual disease-associated variants.

Similar content being viewed by others

References

  1. Polderman, T.J.C. et al. Meta-analysis of the heritability of human traits based on fifty years of twin studies. Nat. Genet. 47, 702–709 (2015).

    CAS  PubMed  Google Scholar 

  2. Sullivan, P.F., Daly, M.J. & O'Donovan, M. Genetic architectures of psychiatric disorders: the emerging picture and its implications. Nat. Rev. Genet. 13, 537–551 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Geschwind, D.H. & Flint, J. Genetics and genomics of psychiatric disease. Science 349, 1489–1494 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Gratten, J., Wray, N.R., Keller, M.C. & Visscher, P.M. Large-scale genomics unveils the genetic architecture of psychiatric disorders. Nat. Neurosci. 17, 782–790 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. McCarroll, S.A., Feng, G. & Hyman, S.E. Genome-scale neurogenetics: methodology and meaning. Nat. Neurosci. 17, 756–763 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Kendler, K.S. What psychiatric genetics has taught us about the nature of psychiatric illness and what is left to learn. Mol. Psychiatry 18, 1058–1066 (2013).

    CAS  PubMed  Google Scholar 

  7. Sham, P.C. & Purcell, S.M. Statistical power and significance testing in large-scale genetic studies. Nat. Rev. Genet. 15, 335–346 (2014).

    CAS  PubMed  Google Scholar 

  8. Flint, J. & Kendler, K.S. The genetics of major depression. Neuron 81, 484–503 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Hawi, Z. et al. The molecular genetic architecture of attention deficit hyperactivity disorder. Mol. Psychiatry 20, 289–297 (2015).

    CAS  PubMed  Google Scholar 

  10. Browne, H.A., Gair, S.L., Scharf, J.M. & Grice, D.E. Genetics of obsessive-compulsive disorder and related disorders. Psychiatr. Clin. North Am. 37, 319–335 (2014).

    PubMed  PubMed Central  Google Scholar 

  11. Manolio, T.A. et al. Finding the missing heritability of complex diseases. Nature 461, 747–753 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

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

  13. 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 

  14. Sanders, S.J. et al. Insights into autism spectrum disorder genomic architecture and biology from 71 risk loci. Neuron 87, 1215–1233 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. 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 

  16. Psychiatric GWAS Consortium Bipolar Disorder Working Group. Large-scale genome-wide association analysis of bipolar disorder identifies a new susceptibility locus near ODZ4. Nat. Genet. 43, 977–983 (2011).

  17. Tammimies, K. et al. Molecular diagnostic yield of chromosomal microarray analysis and whole-exome sequencing in children with autism spectrum disorder. J. Am. Med. Assoc. 314, 895–903 (2015).

    CAS  Google Scholar 

  18. Samocha, K.E. et al. A framework for the interpretation of de novo mutation in human disease. Nat. Genet. 46, 944–950 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. MacArthur, D.G. et al. A systematic survey of loss-of-function variants in human protein-coding genes. Science 335, 823–828 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. MacArthur, D.G. et al. Guidelines for investigating causality of sequence variants in human disease. Nature 508, 469–476 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Sauna, Z.E. & Kimchi-Sarfaty, C. Understanding the contribution of synonymous mutations to human disease. Nat. Rev. Genet. 12, 683–691 (2011).

    CAS  PubMed  Google Scholar 

  22. Cooper, G.M. & Shendure, J. Needles in stacks of needles: finding disease-causal variants in a wealth of genomic data. Nat. Rev. Genet. 12, 628–640 (2011).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Robinson, E.B. et al. Genetic risk for autism spectrum disorders and neuropsychiatric variation in the general population. Nat. Genet. 48, 552–555 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Akbarian, S. et al. The PsychENCODE project. Nat. Neurosci. 18, 1707–1712 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Petersen, A. et al. Evaluating methods for combining rare variant data in pathway-based tests of genetic association. BMC Proc. 5 (Suppl. 9), S48 (2011).

    PubMed  PubMed Central  Google Scholar 

  27. de Leeuw, C.A., Neale, B.M., Heskes, T. & Posthuma, D. The statistical properties of gene-set analysis. Nat. Rev. Genet. 17, 353–364 (2016).

    CAS  PubMed  Google Scholar 

  28. Malhotra, D. & Sebat, J. CNVs: harbingers of a rare variant revolution in psychiatric genetics. Cell 148, 1223–1241 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. de la Torre-Ubieta, L., Won, H., Stein, J.L. & Geschwind, D.H. Advancing the understanding of autism disease mechanisms through genetics. Nat. Med. 22, 345–361 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Zhou, Y. et al. Mice with Shank3 mutations associated with ASD and schizophrenia display both shared and distinct defects. Neuron 89, 147–162 (2016).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Hiroi, N. et al. Copy number variation at 22q11.2: from rare variants to common mechanisms of developmental neuropsychiatric disorders. Mol. Psychiatry 18, 1153–1165 (2013).

    CAS  PubMed  Google Scholar 

  33. Cook, E.H. Jr. et al. Autism or atypical autism in maternally but not paternally derived proximal 15q duplication. Am. J. Hum. Genet. 60, 928–934 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Tansey, K.E. et al. Common alleles contribute to schizophrenia in CNV carriers. Mol. Psychiatry 21, 1085–1089 (2016).

    CAS  PubMed  Google Scholar 

  35. Bernier, R. et al. Disruptive CHD8 mutations define a subtype of autism early in development. Cell 158, 263–276 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Stefansson, H. et al. CNVs conferring risk of autism or schizophrenia affect cognition in controls. Nature 505, 361–366 (2014).

    CAS  PubMed  Google Scholar 

  37. Maillard, A.M. et al. The 16p11.2 locus modulates brain structures common to autism, schizophrenia and obesity. Mol. Psychiatry 20, 140–147 (2015).

    CAS  PubMed  Google Scholar 

  38. Sawcer, S. et al. Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature 476, 214–219 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. 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).

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

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Visel, A., Rubin, E.M. & Pennacchio, L.A. Genomic views of distant-acting enhancers. Nature 461, 199–205 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Maurano, M.T. et al. Systematic localization of common disease-associated variation in regulatory DNA. Science 337, 1190–1195 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Schaub, M.A., Boyle, A.P., Kundaje, A., Batzoglou, S. & Snyder, M. Linking disease associations with regulatory information in the human genome. Genome Res. 22, 1748–1759 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Nord, A.S., Pattabiraman, K., Visel, A. & Rubenstein, J.L.R. Genomic perspectives of transcriptional regulation in forebrain development. Neuron 85, 27–47 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Ernst, J. et al. Mapping and analysis of chromatin state dynamics in nine human cell types. Nature 473, 43–49 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

  48. Sanyal, A., Lajoie, B.R., Jain, G. & Dekker, J. The long-range interaction landscape of gene promoters. Nature 489, 109–113 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Whalen, S., Truty, R.M. & Pollard, K.S. Enhancer-promoter interactions are encoded by complex genomic signatures on looping chromatin. Nat. Genet. 48, 488–496 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Lieberman-Aiden, E. et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science 326, 289–293 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhang, Y. et al. Chromatin connectivity maps reveal dynamic promoter-enhancer long-range associations. Nature 504, 306–310 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Esslinger, C. et al. Neural mechanisms of a genome-wide supported psychosis variant. Science 324, 605 (2009).

    CAS  PubMed  Google Scholar 

  53. Price, A.L., Spencer, C.C.A. & Donnelly, P. Progress and promise in understanding the genetic basis of common diseases. Proc. Biol. Sci. 282, 20151684 (2015).

    PubMed  PubMed Central  Google Scholar 

  54. Wray, N.R., Goddard, M.E. & Visscher, P.M. Prediction of individual genetic risk of complex disease. Curr. Opin. Genet. Dev. 18, 257–263 (2008).

    CAS  PubMed  Google Scholar 

  55. Kendler, K.S., Lönn, S.L., Sundquist, J. & Sundquist, K. Smoking and schizophrenia in population cohorts of Swedish women and men: a prospective co-relative control study. Am. J. Psychiatry 172, 1092–1100 (2015).

    PubMed  PubMed Central  Google Scholar 

  56. French, L. et al. Early cannabis use, polygenic risk score for schizophrenia and brain maturation in adolescence. JAMA Psychiatry 72, 1002–1011 (2015).

    PubMed  PubMed Central  Google Scholar 

  57. Bulik-Sullivan, B.K. et al. LD score regression distinguishes confounding from polygenicity in genome-wide association studies. Nat. Genet. 47, 291–295 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Bulik-Sullivan, B. et al. An atlas of genetic correlations across human diseases and traits. Nat. Genet. 47, 1236–1241 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Flint, J., Timpson, N. & Munafò, M. Assessing the utility of intermediate phenotypes for genetic mapping of psychiatric disease. Trends Neurosci. 37, 733–741 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Finucane, H.K. et al. Partitioning heritability by functional annotation using genome-wide association summary statistics. Nat. Genet. 47, 1228–1235 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Euesden, J., Lewis, C.M. & O'Reilly, P.F. PRSice: Polygenic Risk Score software. Bioinformatics 31, 1466–1468 (2015).

    CAS  PubMed  Google Scholar 

  62. Pasaniuc, B. & Price, A.L. Dissecting the genetics of complex traits using summary association statistics. Preprint at bioRxiv http://dx.doi.org/10.1101/072934 (2016).

  63. Franke, B. et al. Genetic influences on schizophrenia and subcortical brain volumes: large-scale proof of concept. Nat. Neurosci. 19, 420–431 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Vassos, E. et al. An examination of polygenic score risk prediction in individuals with first episode psychosis. Biol. Psychiatry http://dx.doi.org/10.1016/j.biopsych.2016.06.028 (2016).

  65. Ruderfer, D.M. et al. Polygenic dissection of diagnosis and clinical dimensions of bipolar disorder and schizophrenia. Mol. Psychiatry 19, 1017–1024 (2014).

    CAS  PubMed  Google Scholar 

  66. Cleynen, I. et al. Inherited determinants of Crohn's disease and ulcerative colitis phenotypes: a genetic association study. Lancet 387, 156–167 (2016).

    PubMed  PubMed Central  Google Scholar 

  67. Constantino, J.N. & Todd, R.D. Intergenerational transmission of subthreshold autistic traits in the general population. Biol. Psychiatry 57, 655–660 (2005).

    PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Kang, H.J. et al. Spatio-temporal transcriptome of the human brain. Nature 478, 483–489 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Jaffe, A.E. et al. Mapping DNA methylation across development, genotype and schizophrenia in the human frontal cortex. Nat. Neurosci. 19, 40–47 (2016).

    CAS  PubMed  Google Scholar 

  71. Parikshak, N.N. et al. Integrative functional genomic analyses implicate specific molecular pathways and circuits in autism. Cell 155, 1008–1021 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. 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 

  73. 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 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Bakken, T.E. et al. A comprehensive transcriptional map of primate brain development. Nature 535, 367–375 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Bernard, A. et al. Transcriptional architecture of the primate neocortex. Neuron 73, 1083–1099 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. 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 

  78. Darmanis, S. et al. A survey of human brain transcriptome diversity at the single cell level. Proc. Natl. Acad. Sci. USA 112, 7285–7290 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Xu, X., Wells, A.B., O'Brien, D.R., Nehorai, A. & Dougherty, J.D. Cell type-specific expression analysis to identify putative cellular mechanisms for neurogenetic disorders. J. Neurosci. 34, 1420–1431 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. O'Dushlaine, C. et al. Psychiatric genome-wide association study analyses implicate neuronal, immune and histone pathways. Nat. Neurosci. 18, 199–209 (2015).

    CAS  Google Scholar 

  81. 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 

  82. Gilman, S.R. et al. Diverse types of genetic variation converge on functional gene networks involved in schizophrenia. Nat. Neurosci. 15, 1723–1728 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. O'Roak, B.J. et al. Sporadic autism exomes reveal a highly interconnected protein network of de novo mutations. Nature 485, 246–250 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Neale, B.M. et al. Patterns and rates of exonic de novo mutations in autism spectrum disorders. Nature 485, 242–245 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Li, J. et al. Identification of human neuronal protein complexes reveals biochemical activities and convergent mechanisms of action in autism spectrum disorders. Cell Syst. 1, 361–374 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Dimas, A.S. et al. Common regulatory variation impacts gene expression in a cell type-dependent manner. Science 325, 1246–1250 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. 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 

  88. Fromer, M. et al. Gene expression elucidates functional impact of polygenic risk for schizophrenia. Preprint at bioRxiv http://dx.doi.org/10.1101/052209 (2016).

  89. 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 

  90. 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 

  91. Mackay, T.F.C., Stone, E.A. & Ayroles, J.F. The genetics of quantitative traits: challenges and prospects. Nat. Rev. Genet. 10, 565–577 (2009).

    CAS  PubMed  Google Scholar 

  92. Singh, T. et al. Rare loss-of-function variants in SETD1A are associated with schizophrenia and developmental disorders. Nat. Neurosci. 19, 571–577 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Hannon, E. et al. Methylation QTLs in the developing brain and their enrichment in schizophrenia risk loci. Nat. Neurosci. 19, 48–54 (2016).

    CAS  PubMed  Google Scholar 

  94. del Rosario, R.C.-H. et al. Sensitive detection of chromatin-altering polymorphisms reveals autoimmune disease mechanisms. Nat. Methods 12, 458–464 (2015).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  96. Pas¸ca, A.M. et al. Functional cortical neurons and astrocytes from human pluripotent stem cells in 3D culture. Nat. Methods 12, 671–678 (2015).

    Google Scholar 

  97. Dolmetsch, R. & Geschwind, D.H. The human brain in a dish: the promise of iPSC-derived neurons. Cell 145, 831–834 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. 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 

  99. Seok, J. et al. Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc. Natl. Acad. Sci. USA 110, 3507–3512 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Konopka, G. et al. Human-specific transcriptional networks in the brain. Neuron 75, 601–617 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Kwan, K.Y. et al. Species-dependent posttranscriptional regulation of NOS1 by FMRP in the developing cerebral cortex. Cell 149, 899–911 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Geschwind, D.H. & Rakic, P. Cortical evolution: judge the brain by its cover. Neuron 80, 633–647 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Hamilton, S.M. et al. Fmr1 and Nlgn3 knockout rats: novel tools for investigating autism spectrum disorders. Behav. Neurosci. 128, 103–109 (2014).

    CAS  PubMed  Google Scholar 

  104. Liu, Z. et al. Autism-like behaviours and germline transmission in transgenic monkeys overexpressing MeCP2. Nature 530, 98–102 (2016).

    CAS  PubMed  Google Scholar 

  105. Kaiser, T. & Feng, G. Modeling psychiatric disorders for developing effective treatments. Nat. Med. 21, 979–988 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Jeste, S.S. & Geschwind, D.H. Clinical trials for neurodevelopmental disorders: at a therapeutic frontier. Sci. Transl. Med. 8, 321fs1 (2016).

    PubMed  Google Scholar 

  107. Dockendorff, T.C. et al. Drosophila lacking dfmr1 activity show defects in circadian output and fail to maintain courtship interest. Neuron 34, 973–984 (2002).

    CAS  PubMed  Google Scholar 

  108. Hoffman, E.J. et al. Estrogens suppress a behavioral phenotype in zebrafish mutants of the autism risk gene, CNTNAP2. Neuron 89, 725–733 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Portmann, T. et al. Behavioral abnormalities and circuit defects in the basal ganglia of a mouse model of 16p11.2 deletion syndrome. Cell Reports 7, 1077–1092 (2014).

    CAS  PubMed  Google Scholar 

  110. 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 

  111. Pas¸ca, S.P. et al. Using iPSC-derived neurons to uncover cellular phenotypes associated with Timothy syndrome. Nat. Med. 17, 1657–1662 (2011).

    Google Scholar 

  112. Mariani, J. et al. FOXG1-dependent dysregulation of GABA/glutamate neuron differentiation in autism spectrum disorders. Cell 162, 375–390 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Patwardhan, R.P. et al. Massively parallel functional dissection of mammalian enhancers in vivo. Nat. Biotechnol. 30, 265–270 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Mei, Y. et al. Adult restoration of Shank3 expression rescues selective autistic-like phenotypes. Nature 530, 481–484 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Stein, J.L. et al. A quantitative framework to evaluate modeling of cortical development by neural stem cells. Neuron 83, 69–86 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Hyman, S.E. Revolution stalled. Sci. Transl. Med. 4, 155cm11 (2012).

    PubMed  Google Scholar 

  117. Plenge, R.M., Scolnick, E.M. & Altshuler, D. Validating therapeutic targets through human genetics. Nat. Rev. Drug Discov. 12, 581–594 (2013).

    CAS  PubMed  Google Scholar 

  118. Hou, L. et al. Genetic variants associated with response to lithium treatment in bipolar disorder: a genome-wide association study. Lancet 387, 1085–1093 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Goldstein, J.I. et al. Clozapine-induced agranulocytosis is associated with rare HLA-DQB1 and HLA-B alleles. Nat. Commun. 5, 4757 (2014).

    CAS  PubMed  Google Scholar 

  120. Lajonchere, C.M. Changing the landscape of autism research: the autism genetic resource exchange. Neuron 68, 187–191 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Félix, M.-A. & Barkoulas, M. Pervasive robustness in biological systems. Nat. Rev. Genet. 16, 483–496 (2015).

    PubMed  Google Scholar 

  122. Klei, L. et al. Common genetic variants, acting additively, are a major source of risk for autism. Mol. Autism 3, 9 (2012).

    PubMed  PubMed Central  Google Scholar 

  123. Gaugler, T. et al. Most genetic risk for autism resides with common variation. Nat. Genet. 46, 881–885 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Nelson, M.R. et al. The support of human genetic evidence for approved drug indications. Nat. Genet. 47, 856–860 (2015).

    CAS  PubMed  Google Scholar 

  125. Teslovich, T.M. et al. Biological, clinical and population relevance of 95 loci for blood lipids. Nature 466, 707–713 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Bush, W.S., Oetjens, M.T. & Crawford, D.C. Unravelling the human genome-phenome relationship using phenome-wide association studies. Nat. Rev. Genet. 17, 129–145 (2016).

    CAS  PubMed  Google Scholar 

  127. Kohane, I.S. Using electronic health records to drive discovery in disease genomics. Nat. Rev. Genet. 12, 417–428 (2011).

    CAS  PubMed  Google Scholar 

  128. Nelson, M.R. et al. The genetics of drug efficacy: opportunities and challenges. Nat. Rev. Genet. 17, 197–206 (2016).

    CAS  PubMed  Google Scholar 

  129. Rastegar-Mojarad, M., Ye, Z., Kolesar, J.M., Hebbring, S.J. & Lin, S.M. Opportunities for drug repositioning from phenome-wide association studies. Nat. Biotechnol. 33, 342–345 (2015).

    CAS  PubMed  Google Scholar 

  130. Li, J. et al. A survey of current trends in computational drug repositioning. Brief. Bioinform. 17, 2–12 (2016).

    PubMed  Google Scholar 

  131. Ruderfer, D.M. et al. Polygenic overlap between schizophrenia risk and antipsychotic response: a genomic medicine approach. Lancet Psychiatry 3, 350–357 (2016).

    PubMed  PubMed Central  Google Scholar 

  132. Lamb, J. et al. The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. Science 313, 1929–1935 (2006).

    CAS  PubMed  Google Scholar 

  133. Lichtenstein, P. et al. Common genetic determinants of schizophrenia and bipolar disorder in Swedish families: a population-based study. Lancet 373, 234–239 (2009).

    CAS  PubMed  Google Scholar 

  134. Lee, S.H. et al. Genetic relationship between five psychiatric disorders estimated from genome-wide SNPs. Nat. Genet. 45, 984–994 (2013).

    CAS  PubMed  Google Scholar 

  135. Reichenberg, A. et al. Discontinuity in the genetic and environmental causes of the intellectual disability spectrum. Proc. Natl. Acad. Sci. USA 113, 1098–1103 (2016).

    CAS  PubMed  Google Scholar 

  136. Gilissen, C. et al. Genome sequencing identifies major causes of severe intellectual disability. Nature 511, 344–347 (2014).

    CAS  PubMed  Google Scholar 

  137. Plomin, R., Haworth, C.M., Meaburn, E.L., Price, T.S. & Davis, O.S. Common DNA markers can account for more than half of the genetic influence on cognitive abilities. Psychol. Sci. 24, 562–568 (2013).

    PubMed  Google Scholar 

  138. Ridge, P.G. et al. Assessment of the genetic variance of late-onset Alzheimer's disease. Neurobiol. Aging 41, 200.e13–e20 (2016).

    PubMed  PubMed Central  Google Scholar 

  139. Tenesa, A. & Haley, C.S. The heritability of human disease: estimation, uses and abuses. Nat. Rev. Genet. 14, 139–149 (2013).

    CAS  PubMed  Google Scholar 

  140. 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 

  141. Mutsuddi, M. et al. Analysis of high-resolution HapMap of DTNBP1 (dysbindin) suggests no consistency between reported common variant associations and schizophrenia. Am. J. Hum. Genet. 79, 903–909 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Spielman, R.S. et al. Common genetic variants account for differences in gene expression among ethnic groups. Nat. Genet. 39, 226–231 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Akey, J.M., Biswas, S., Leek, J.T. & Storey, J.D. On the design and analysis of gene expression studies in human populations. Nat. Genet. 39, 807–808 author reply 808–809 (2007).

    CAS  PubMed  Google Scholar 

  144. Sebastiani, P. et al. Genetic signatures of exceptional longevity in humans. Science 2010, 1190532 (2010).

    Google Scholar 

  145. Skafidas, E. et al. Predicting the diagnosis of autism spectrum disorder using gene pathway analysis. Mol. Psychiatry 19, 504–510 (2014).

    CAS  PubMed  Google Scholar 

  146. Belgard, T.G., Jankovic, I., Lowe, J.K. & Geschwind, D.H. Population structure confounds autism genetic classifier. Mol. Psychiatry 19, 405–407 (2014).

    CAS  PubMed  Google Scholar 

  147. Arnedo, J. et al. Uncovering the hidden risk architecture of the schizophrenias: confirmation in three independent genome-wide association studies. Am. J. Psychiatry 172, 139–153 (2015).

    PubMed  Google Scholar 

  148. Wang, Z. et al. Nuclear receptor NR1H3 in familial multiple sclerosis. Neuron 90, 948–954 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Cotsapas, C. NR1H3 p.Arg415Gln is not associated to multiple sclerosis risk. Preprint at bioRxiv http://dx.doi.org/10.1101/061366 (2016).

Download references

Acknowledgements

The authors thank A. Gordon, L. Perez-Cano, G. Ramaswami, E. Ruzzo, J. Rexach, G. Morris, and members of the Geschwind laboratory for discussions. This work was supported by US National Institute of Health (NIH) grants 5R01MH094714 (D.H.G.), 5P50MH106438 (D.H.G.), 1R01MH109912 (D.H.G.), and F30MH099886 (N.N.P.). This work was also supported by the Glenn/AFAR Postdoctoral Fellowship (20145357, H.W.), Basic Science Research Program through the National Research Foundation of Korea (2013024227, H.W.), and the UCLA Medical Scientist Training Program (N.N.P.). D.H.G. is a scientific advisor to Ovid Pharmaceuticals.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Daniel H Geschwind.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Table 1 (PDF 63 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gandal, M., Leppa, V., Won, H. et al. The road to precision psychiatry: translating genetics into disease mechanisms. Nat Neurosci 19, 1397–1407 (2016). https://doi.org/10.1038/nn.4409

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4409

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