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.

Genetic insights into the neurodevelopmental origins of schizophrenia

Key Points

  • Schizophrenia is a severe neuropsychiatric disorder with an unknown causative pathophysiology.

  • Genome-wide association studies applied to schizophrenia have successfully identified discrete risk variants of common and rare frequency, but the functional characterization and neurobiological import of these loci remain to be elucidated.

  • The neurodevelopmental model provides a translational context for schizophrenia genetics, positing that schizophrenia genetic and epigenetic risk factors converge on early brain development to perturb neurodevelopmental trajectories.

  • Post-mortem studies of brain gene expression and DNA methylation suggest that risk factors for schizophrenia, both genetic and epigenetic variations that leave marks in the adult brain, occur principally during early brain development rather than during the tumultuous period of late adolescence or early adulthood, when the diagnosis is typically made.

  • The diverse and protean effects of genetic and epigenetic risk of schizophrenia on brain development may be parsimoniously conceptualized as introducing developmental 'noise' — various subtle perturbations, including those to early brain circuits and synaptic organization — analogous to alterations in autism and intellectual disability but with lesser 'noise' burden or more amenable to compensation.

  • Schizophrenia risk loci should be functionally characterized (spatially and temporally) in relevant neurodevelopmental models, including post-mortem brain across developmental stages and placenta and stem cell models.

Abstract

Schizophrenia is a severe neuropsychiatric disorder with a longstanding history of neurobiological investigation. Although the underlying causal mechanisms remain unknown, early neurodevelopmental events have been implicated in pathogenesis, initially by epidemiological and circumstantial data but more recently by brain-specific molecular and genetic findings. Notably, genomic research has recently uncovered discrete risk variants and risk loci associated with schizophrenia, with the potential to elucidate disease mechanisms. This Review revisits the neurodevelopmental model of schizophrenia from a current genetics perspective, delineating the complex genetic basis of the disorder and highlighting gene expression and epigenetic analyses of post-mortem cortical tissue that suggest that early brain development mediates genetic risk associated with schizophrenia. Future functional genomics investigations will accordingly need to characterize schizophrenia risk loci in relevant neurodevelopmental models.

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 epigenetic risk factors for schizophrenia converge on neurodevelopment.

References

  1. Jablensky, A., Kirkbride, J. B. & Jones, P. B. in Schizophrenia 3rd edn Ch. 10 (eds Weinberger, D. & Harrison, P. J.) (Blackwell, 2011).

    Google Scholar 

  2. Jaspers, K. General Psychopathology [German] (Manchester Univ. Press, 1959).

    Google Scholar 

  3. American Psychiatric Association. Diagnostic and statistical manual of mental disorders 5th edn (American Psychiatric Publishing, 2013).

  4. Bleuler, E. Dementia Praecox or the Group of Schizophrenias (International Universities Press,1950).

    Google Scholar 

  5. Andreasen, N. C. in Schizophrenia 3rd edn Ch. 1 (eds Weinberger, D. & Harrison, P. J.) (Blackwell, 2011).

    Google Scholar 

  6. Sherrington, R. et al. Localization of a susceptibility locus for schizophrenia on chromosome 5. Nature 336, 164–167 (1988).

    Article  CAS  PubMed  Google Scholar 

  7. [No authors listed.] Where next with psychiatric illness? Nature 336, 95–96 (1988).

  8. Harrison, P., Lewis, D. E. & Kleinman, J. E. in Schizophrenia 3rd edn Ch. 18 (eds Weinberger, D. & Harrison, P. J.) (Blackwell, 2011).

    Google Scholar 

  9. Harrison, P. J. Postmortem studies in schizophrenia. Dialogues Clin. Neurosci. 2, 349–357 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Weinberger, D. R. & Radulescu, E. Finding the elusive psychiatric “lesion” with 21st-century neuroanatomy: a note of caution. Am. J. Psychiatry 173, 27–33 (2016).

    Article  PubMed  Google Scholar 

  11. Johnstone, E. C., Crow, T. J., Frith, C. D., Husband, J. & Kreel, L. Cerebral ventricular size and cognitive impairment in chronic schizophrenia. Lancet 2, 924–926 (1976).

    Article  CAS  PubMed  Google Scholar 

  12. Adriano, F., Caltagirone, C. & Spalletta, G. Hippocampal volume reduction in first-episode and chronic schizophrenia: a review and meta-analysis. Neuroscientist 18, 180–200 (2012).

    Article  PubMed  Google Scholar 

  13. Lieberman, J. A. Is schizophrenia a neurodegenerative disorder? A clinical and neurobiological perspective. Biol. Psychiatry 46, 729–739 (1999).

    Article  CAS  PubMed  Google Scholar 

  14. Miller, B. J., Buckley, P., Seabolt, W., Mellor, A. & Kirkpatrick, B. Meta-analysis of cytokine alterations in schizophrenia: clinical status and antipsychotic effects. Biol. Psychiatry 70, 663–671 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Volk, D. W. et al. Molecular mechanisms and timing of cortical immune activation in schizophrenia. Am. J. Psychiatry 172, 1112–1121 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Bloomfield, P. S. et al. Microglial activity in people at ultra high risk of psychosis and in schizophrenia: an [11C]PBR28 PET brain imaging study. Am. J. Psychiatry 173, 44–52 (2016).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Birnbaum, R. et al. Investigating the neuroimmunogenic architecture of schizophrenia. Mol. Psychiatry http://dx.doi.org/10.1038/mp.2017.89 (2017).

  19. MacDonald, M. L. et al. Selective loss of smaller spines in schizophrenia. Am. J. Psychiatry 174, 586–594 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Carlsson, A. & Lindqvist, M. Effect of chlorpromazine or haloperidol on formation of 3-methoxytyramine and normetanephrine in mouse brain. Acta Pharmacol. Toxicol. (Copenh.) 20, 140–144 (1963).

    Article  CAS  Google Scholar 

  21. Howes, O. D. & Kapur, S. The dopamine hypothesis of schizophrenia: version III — the final common pathway. Schizophr. Bull. 35, 549–562 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Abi-Dargham, A. & Grace, A. A. in Schizophrenia 3rd edn Ch. 20 (eds Weinberger, D. & Harrison, P. J.) (Blackwell, 2011).

    Google Scholar 

  23. Weinberger, D. R., Berman, K. F. & Zec, R. F. Physiologic dysfunction of dorsolateral prefrontal cortex in schizophrenia. I. Regional cerebral blood flow evidence. Arch. Gen. Psychiatry 43, 114–124 (1986). This article identifies functional changes in the DLPFC during a working-memory task in medication-free schizophrenia patients, positing a region-specific localization of schizophrenia pathophysiology.

    Article  CAS  PubMed  Google Scholar 

  24. Goldman-Rakic, P. S., Selemon, L. D. & Schwartz, M. L. Dual pathways connecting the dorsolateral prefrontal cortex with the hippocampal formation and parahippocampal cortex in the rhesus monkey. Neuroscience 12, 719–743 (1984).

    Article  CAS  PubMed  Google Scholar 

  25. Anticevic, A., Repovs, G., Krystal, J. H. & Barch, D. M. A broken filter: prefrontal functional connectivity abnormalities in schizophrenia during working memory interference. Schizophr. Res. 141, 8–14 (2012).

    Article  PubMed  Google Scholar 

  26. Callicott, J. H. et al. Physiological dysfunction of the dorsolateral prefrontal cortex in schizophrenia revisited. Cereb. Cortex 10, 1078–1092 (2000).

    Article  CAS  PubMed  Google Scholar 

  27. Lipska, B. K., Jaskiw, G. E. & Weinberger, D. R. Postpubertal emergence of hyperresponsiveness to stress and to amphetamine after neonatal excitotoxic hippocampal damage: a potential animal model of schizophrenia. Neuropsychopharmacology 9, 67–75 (1993).

    Article  CAS  PubMed  Google Scholar 

  28. Goldman-Rakic, P. S. Regional and cellular fractionation of working memory. Proc. Natl Acad. Sci. USA 93, 13473–13480 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Heckers, S. Neuroimaging studies of the hippocampus in schizophrenia. Hippocampus 11, 520–528 (2001).

    Article  CAS  PubMed  Google Scholar 

  30. Heckers, S. & Konradi, C. Hippocampal neurons in schizophrenia. J. Neural Transm. (Vienna) 109, 891–905 (2002).

    Article  CAS  Google Scholar 

  31. Weinberger, D. R. Implications of normal brain development for the pathogenesis of schizophrenia. Arch. Gen. Psychiatry 44, 660–669 (1987). This article introduces and expounds the classic neurodevelopmental model of schizophrenia.

    Article  CAS  PubMed  Google Scholar 

  32. Murray, R. M. & Lewis, S. W. Is schizophrenia a neurodevelopmental disorder? Br. Med. J. (Clin. Res. Ed.) 296, 63 (1988).

    Article  CAS  Google Scholar 

  33. Feinberg, I. Schizophrenia: caused by a fault in programmed synaptic elimination during adolescence? J. Psychiatr. Res. 17, 319–334 (1982).

    Article  PubMed  Google Scholar 

  34. Rapoport, J. L., Addington, A. M., Frangou, S. & Psych, M. R. The neurodevelopmental model of schizophrenia: update 2005. Mol. Psychiatry 10, 434–449 (2005).

    Article  CAS  PubMed  Google Scholar 

  35. Lewis, D. A. & Levitt, P. Schizophrenia as a disorder of neurodevelopment. Annu. Rev. Neurosci. 25, 409–432 (2002).

    Article  CAS  PubMed  Google Scholar 

  36. Krystal, J. H. et al. Impaired tuning of neural ensembles and the pathophysiology of schizophrenia: a translational and computational neuroscience perspective. Biol. Psychiatry 81, 874–885 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Stiles, J. & Jernigan, T. L. The basics of brain development. Neuropsychol. Rev. 20, 327–348 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Johnson, M. H. Functional brain development in humans. Nat. Rev. Neurosci. 2, 475–483 (2001).

    Article  CAS  PubMed  Google Scholar 

  39. Fahrbach, S. E. Developmental Neuroscience: A Concise Introduction (Princeton Univ. Press, 2013).

    Google Scholar 

  40. Stiles, J. The Fundementals Of Brain Development: Integrating Nature And Nurture (Harvard Univ. Press, 2008).

    Google Scholar 

  41. Catts, V. S. et al. Rethinking schizophrenia in the context of normal neurodevelopment. Front. Cell. Neurosci. 7, 60 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Bercury, K. K. & Macklin, W. B. Dynamics and mechanisms of CNS myelination. Dev. Cell 32, 447–458 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Buss, R. R., Sun, W. & Oppenheim, R. W. Adaptive roles of programmed cell death during nervous system development. Annu. Rev. Neurosci. 29, 1–35 (2006).

    Article  CAS  PubMed  Google Scholar 

  44. Tau, G. Z. & Peterson, B. S. Normal development of brain circuits. Neuropsychopharmacology 35, 147–168 (2010).

    Article  PubMed  Google Scholar 

  45. Marin, O., Valiente, M., Ge, X. & Tsai, L. H. Guiding neuronal cell migrations. Cold Spring Harb. Perspect. Biol. 2, a001834 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Gotz, M. & Huttner, W. B. The cell biology of neurogenesis. Nat. Rev. Mol. Cell Biol. 6, 777–788 (2005).

    Article  CAS  PubMed  Google Scholar 

  47. Rakic, P. Specification of cerebral cortical areas. Science 241, 170–176 (1988).

    Article  CAS  PubMed  Google Scholar 

  48. Echevarria, D., Vieira, C., Gimeno, L. & Martinez, S. Neuroepithelial secondary organizers and cell fate specification in the developing brain. Brain Res. Brain Res. Rev. 43, 179–191 (2003).

    Article  CAS  PubMed  Google Scholar 

  49. Grove, E. A. & Fukuchi-Shimogori, T. Generating the cerebral cortical area map. Annu. Rev. Neurosci. 26, 355–380 (2003).

    Article  CAS  PubMed  Google Scholar 

  50. Greenough, W. T. & Chang, F. F. in Cerebral Cortex (eds Peters, A. & Jones, E. G.) 391–440 (Plenum, 1988).

    Book  Google Scholar 

  51. Weinberger, D. R. & Levitt, P. in Schizophrenia 3rd edn Ch. 19 (eds Weinberger, D. & Harrison, P. J.) (Blackwell, 2011).

    Google Scholar 

  52. Lipska, B. K. & Weinberger, D. R. Delayed effects of neonatal hippocampal damage on haloperidol-induced catalepsy and apomorphine-induced stereotypic behaviors in the rat. Brain Res. Dev. Brain Res. 75, 213–222 (1993).

    Article  CAS  PubMed  Google Scholar 

  53. Lipska, B. K. & Weinberger, D. R. To model a psychiatric disorder in animals: schizophrenia as a reality test. Neuropsychopharmacology 23, 223–239 (2000).

    Article  CAS  PubMed  Google Scholar 

  54. Floresco, S. B., Geyer, M. A., Gold, L. H. & Grace, A. A. Developing predictive animal models and establishing a preclinical trials network for assessing treatment effects on cognition in schizophrenia. Schizophr. Bull. 31, 888–894 (2005).

    Article  PubMed  Google Scholar 

  55. Saunders, R. C., Kolachana, B. S., Bachevalier, J. & Weinberger, D. R. Neonatal lesions of the medial temporal lobe disrupt prefrontal cortical regulation of striatal dopamine. Nature 393, 169–171 (1998).

    Article  CAS  PubMed  Google Scholar 

  56. Sullivan, P. F., Kendler, K. S. & Neale, M. C. Schizophrenia as a complex trait: evidence from a meta-analysis of twin studies. Arch. Gen. Psychiatry 60, 1187–1192 (2003).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  58. Riley, B. & Kendler, K. in Schizophrenia 3rd edn Ch. 12 (eds Weinberger, D. & Harrison, P. J.) (Blackwell, 2011).

    Google Scholar 

  59. Schizophrenia Working Group of the Psychiatric Genomics Consortium. Biological insights from 108 schizophrenia-associated genetic loci. Nature 511, 421–427 (2014). This report by the PGC is the largest meta-analysis to date of SNP common variants associated with schizophrenia based on a genome-wide association case–control study design.

  60. Lee, S. H., Goddard, M. E., Wray, N. R. & Visscher, P. M. A better coefficient of determination for genetic profile analysis. Genet. Epidemiol. 36, 214–224 (2012).

    Article  PubMed  Google Scholar 

  61. Marhsall, C. R. et al. Contribution of copy number variants to schizophrenia from a genome-wide study of 41,321 subjects. Nat. Genet. 49, 27–35 (2017). This report by the PGC is the largest meta-analysis to date of CNV rare variants associated with schizophrenia based on a genome-wide association case–control study design.

    Article  CAS  Google Scholar 

  62. Girirajan, S. et al. Phenotypic heterogeneity of genomic disorders and rare copy-number variants. N. Engl. J. Med. 367, 1321–1331 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Grayton, H. M., Fernandes, C., Rujescu, D. & Collier, D. A. Copy number variations in neurodevelopmental disorders. Prog. Neurobiol. 99, 81–91 (2012).

    Article  CAS  PubMed  Google Scholar 

  64. Girirajan, S. & Eichler, E. E. Phenotypic variability and genetic susceptibility to genomic disorders. Hum. Mol. Genet. 19, R176–R187 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  66. Owen, M. J., O'Donovan, M. C., Thapar, A. & Craddock, N. Neurodevelopmental hypothesis of schizophrenia. Br. J. Psychiatry 198, 173–175 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  67. O'Donovan, M. C., Kirov, G. & Owen, M. J. Phenotypic variations on the theme of CNVs. Nat. Genet. 40, 1392–1393 (2008).

    Article  CAS  PubMed  Google Scholar 

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

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Genovese, G. et al. Increased burden of ultra-rare protein-altering variants among 4,877 individuals with schizophrenia. Nat. Neurosci. 19, 1433–1441 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Singh, T. et al. The contribution of rare variants to risk of schizophrenia in individuals with and without intellectual disability. Nat. Genet. 49, 1167–1173 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Koboldt, D. C., Steinberg, K. M., Larson, D. E., Wilson, R. K. & Mardis, E. R. The next-generation sequencing revolution and its impact on genomics. Cell 155, 27–38 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Phillips, P. C. Epistasis — the essential role of gene interactions in the structure and evolution of genetic systems. Nat. Rev. Genet. 9, 855–867 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Weinberger, D. R. Epistasis in schizophrenia genetics: what's missing is not heritability. Schizophr. Res. 160, e2–e3 (2014).

    Article  Google Scholar 

  78. Mackay, T. F. C. Epistasis and quantitative traits: using model organisms to study gene–gene interactions. Nat. Rev. Genet. 15, 22–33 (2014).

    Article  CAS  PubMed  Google Scholar 

  79. Zuk, O., Hechter, E., Sunyaev, S. R. & Lander, E. S. The mystery of missing heritability: genetic interactions create phantom heritability. Proc. Natl Acad. Sci. USA 109, 1193–1198 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Arnsten, A. F. Catecholamine regulation of the prefrontal cortex. J. Psychopharmacol. 11, 151–162 (1997).

    Article  CAS  PubMed  Google Scholar 

  81. Brozoski, T. J., Brown, R. M., Rosvold, H. E. & Goldman, P. S. Cognitive deficit caused by regional depletion of dopamine in prefrontal cortex of rhesus monkey. Science 205, 929–932 (1979).

    Article  CAS  PubMed  Google Scholar 

  82. Horn, T. et al. Mapping of signaling networks through synthetic genetic interaction analysis by RNAi. Nat. Methods 8, 341–346 (2011).

    Article  CAS  PubMed  Google Scholar 

  83. Szappanos, B. et al. An integrated approach to characterize genetic interaction networks in yeast metabolism. Nat. Genet. 43, 656–662 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Lehner, B., Crombie, C., Tischler, J., Fortunato, A. & Fraser, A. G. Systematic mapping of genetic interactions in Caenorhabditis elegans identifies common modifiers of diverse signaling pathways. Nat. Genet. 38, 896–903 (2006).

    Article  CAS  PubMed  Google Scholar 

  85. Nicodemus, K. K. et al. Evidence of statistical epistasis between DISC1, CIT and NDEL1 impacting risk for schizophrenia: biological validation with functional neuroimaging. Hum. Genet. 127, 441–452 (2010).

    Article  CAS  PubMed  Google Scholar 

  86. Nicodemus, K. K. et al. Variability in working memory performance explained by epistasis versus polygenic scores in the ZNF804A pathway. JAMA Psychiatry 71, 778–785 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. 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).

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  90. Li, Y. I. et al. RNA splicing is a primary link between genetic variation and disease. Science 352, 600–604 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Hill, M. J. & Bray, N. J. Evidence that schizophrenia risk variation in the ZNF804A gene exerts its effects during fetal brain development. Am. J. Psychiatry 169, 1301–1308 (2012).

    Article  PubMed  Google Scholar 

  93. Tao, R. et al. Expression of ZNF804A in human brain and alterations in schizophrenia, bipolar disorder, and major depressive disorder: a novel transcript fetally regulated by the psychosis risk variant rs1344706. JAMA Psychiatry 71, 1112–1120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. 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).

  95. Li, M. et al. A human-specific AS3MT isoform and BORCS7 are molecular risk factors in the 10q24.32 schizophrenia-associated locus. Nat. Med. 22, 649–656 (2016).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Birnbaum, R. et al. Investigation of the prenatal expression patterns of 108 schizophrenia-associated genetic loci. Biol. Psychiatry 77, e43–e51 (2015).

    Article  CAS  PubMed  Google Scholar 

  98. Birnbaum, R., Jaffe, A. E., Hyde, T. M., Kleinman, J. E. & Weinberger, D. R. Prenatal expression patterns of genes associated with neuropsychiatric disorders. Am. J. Psychiatry 171, 758–767 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  99. Jenkins, A. K. et al. Neurexin 1 (NRXN1) splice isoform expression during human neocortical development and aging. Mol. Psychiatry 21, 701–706 (2016).

    Article  CAS  PubMed  Google Scholar 

  100. Jaffe, A. et al. Development and genetic regulation of the human cortex transcriptome in schizophrenia. Preprint at https://www.biorxiv.org/content/biorxiv/early/2017/04/05/124321.full.pdf (2017).

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Willsey, A. J. et al. Coexpression networks implicate human midfetal deep cortical projection neurons in the pathogenesis of autism. Cell 155, 997–1007 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Ellis, S. E., Panitch, R., West, A. B. & Arking, D. E. Transcriptome analysis of cortical tissue reveals shared sets of downregulated genes in autism and schizophrenia. Transl Psychiatry 6, e817 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Ursini, G. et al. Placental gene expression mediates the interaction between obstetrical history and genetic risk for schizophrenia. Preprint at https://www.biorxiv.org/content/early/2017/06/07/147207 (2017).

  109. Brennand, K. J. & Gage, F. H. Concise review: the promise of human induced pluripotent stem cell-based studies of schizophrenia. Stem Cells 29, 1915–1922 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Brennand, K. J. et al. Creating patient-specific neural cells for the in vitro study of brain disorders. Stem Cell Reports 5, 933–945 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  111. Ho, S. M., Topol, A. & Brennand, K. J. From “directed differentiation” to “neuronal induction”: modeling neuropsychiatric disease. Biomark. Insights 10 (Suppl. 1), 31–41 (2015).

    PubMed  PubMed Central  Google Scholar 

  112. Brennand, K. J., Landek-Salgado, M. A. & Sawa, A. Modeling heterogeneous patients with a clinical diagnosis of schizophrenia with induced pluripotent stem cells. Biol. Psychiatry 75, 936–944 (2014).

    Article  PubMed  Google Scholar 

  113. Soliman, M. A., Aboharb, F., Zeltner, N. & Studer, L. Pluripotent stem cells in neuropsychiatric disorders. Mol. Psychiatry 22, 1241–1249 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Boyle, E. A., Li, Y. I. & Pritchard, J. K. An expanded view of complex traits: from polygenic to omnigenic. Cell 169, 1177–1186 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Macosko, E. Z. et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161, 1202–1214 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Wei, P. C. et al. Long neural genes harbor recurrent DNA break clusters in neural stem/progenitor cells. Cell 164, 644–655 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Weissman, I. L. & Gage, F. H. Mechanism for somatic brain mosaicism. Cell 164, 593–595 (2016).

    Article  CAS  PubMed  Google Scholar 

  118. Ziegenhain, C. et al. Comparative analysis of single-cell RNA sequencing methods. Mol. Cell 65, 631–643.e4 (2017).

    Article  CAS  PubMed  Google Scholar 

  119. Ramani, V. et al. Massively multiplex single-cell Hi-C. Nat. Methods 14, 263–266 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. McConnell, M. J. et al. Intersection of diverse neuronal genomes and neuropsychiatric disease: The Brain Somatic Mosaicism Network. Science 356, eaal1641 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Kukurba, K. R. & Montgomery, S. B. RNA sequencing and analysis. Cold Spring Harb. Protoc. 2015, 951–969 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  122. Hwang, T. et al. Dynamic regulation of RNA editing in human brain development and disease. Nat. Neurosci. 19, 1093–1099 (2016).

    Article  CAS  PubMed  Google Scholar 

  123. Jaffe, A. E. et al. qSVA framework for RNA quality correction in differential expression analysis. Proc. Natl Acad. Sci. USA 114, 7130–7135 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Sklar, P. in Neurobiology of Mental Illness (eds Charney, D. S., Buxbaum, J. D., Sklar, P. & Nestler, E. J.) 2–246 (Oxford Univ. Press, 2013).

    Google Scholar 

  125. Weinberger, D. R., Glick, I. D. & Klein, D. F. Whither research domain criteria (RDoC)?: the good, the bad, and the ugly. JAMA Psychiatry 72, 1161–1162 (2015).

    Article  PubMed  Google Scholar 

  126. Krystal, J. H. et al. Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans. Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch. Gen. Psychiatry 51, 199–214 (1994).

    Article  CAS  PubMed  Google Scholar 

  127. van Rossum, J. M. The significance of dopamine-receptor blockade for the mechanism of action of neuroleptic drugs. Arch. Int. Pharmacodyn. Ther. 160, 492–494 (1966).

    CAS  PubMed  Google Scholar 

  128. Javitt, D. C. Glutamate and schizophrenia: phencyclidine, N-methyl-d-aspartate receptors, and dopamine-glutamate interactions. Int. Rev. Neurobiol. 78, 69–108 (2007).

    Article  CAS  PubMed  Google Scholar 

  129. Seeman, P., Chau-Wong, M., Tedesco, J. & Wong, K. Brain receptors for antipsychotic drugs and dopamine: direct binding assays. Proc. Natl Acad. Sci. USA 72, 4376–4380 (1975).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Abi-Dargham, A. Dopamine dysfunction in schizophrenia. Schizophr. Res. 160, e6–e7 (2014).

    Article  Google Scholar 

  131. Abi-Dargham, A. Do we still believe in the dopamine hypothesis? New data bring new evidence. Int. J. Neuropsychopharmacol. 7, S1–S5 (2004).

    Article  CAS  PubMed  Google Scholar 

  132. Jaskiw, G. E., Weinberger, D. R. & Crawley, J. N. Microinjection of apomorphine into the prefrontal cortex of the rat reduces dopamine metabolite concentrations in microdialysate from the caudate nucleus. Biol. Psychiatry 29, 703–706 (1991).

    Article  CAS  PubMed  Google Scholar 

  133. Howes, O. D. et al. The nature of dopamine dysfunction in schizophrenia and what this means for treatment. Arch. Gen. Psychiatry 69, 776–786 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Davis, K. L., Kahn, R. S., Ko, G. & Davidson, M. Dopamine in schizophrenia: a review and reconceptualization. Am. J. Psychiatry 148, 1474–1486 (1991).

    Article  CAS  PubMed  Google Scholar 

  135. Coyle, J. T. Glutamate and schizophrenia: beyond the dopamine hypothesis. Cell. Mol. Neurobiol. 26, 365–384 (2006).

    Article  CAS  PubMed  Google Scholar 

  136. Hu, W., MacDonald, M. L., Elswick, D. E. & Sweet, R. A. The glutamate hypothesis of schizophrenia: evidence from human brain tissue studies. Ann. N.Y. Acad. Sci. 1338, 38–57 (2015).

    Article  CAS  PubMed  Google Scholar 

  137. Krystal, J. H. & Moghaddam, B. in Schizophrenia 3rd edn Ch. 21 (eds Weinberger, D. & Harrison, P. J.) (Blackwell, 2011).

    Google Scholar 

  138. Schmidt, M. J. & Mirnics, K. Neurodevelopment, GABA system dysfunction, and schizophrenia. Neuropsychopharmacology 40, 190–206 (2015).

    Article  PubMed  Google Scholar 

  139. Hyde, T. M. et al. Expression of GABA signaling molecules KCC2, NKCC1, and GAD1 in cortical development and schizophrenia. J. Neurosci. 31, 11088–11095 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Mitchell, A. C., Jiang, Y., Peter, C. & Akbarian, S. Transcriptional regulation of GAD1 GABA synthesis gene in the prefrontal cortex of subjects with schizophrenia. Schizophr. Res. 167, 28–34 (2015).

    Article  PubMed  Google Scholar 

  141. Guillozet-Bongaarts, A. L. et al. Altered gene expression in the dorsolateral prefrontal cortex of individuals with schizophrenia. Mol. Psychiatry 19, 478–485 (2014).

    Article  CAS  PubMed  Google Scholar 

  142. Gonzalez-Burgos, G. & Lewis, D. A. NMDA receptor hypofunction, parvalbumin-positive neurons, and cortical gamma oscillations in schizophrenia. Schizophr. Bull. 38, 950–957 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  143. Lewis, D. A. Inhibitory neurons in human cortical circuits: substrate for cognitive dysfunction in schizophrenia. Curr. Opin. Neurobiol. 26, 22–26 (2014).

    Article  CAS  PubMed  Google Scholar 

  144. Curley, A. A. & Lewis, D. A. Cortical basket cell dysfunction in schizophrenia. J. Physiol. 590, 715–724 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Kahn, R. S. et al. Schizophrenia. Nat. Rev. Dis. Primers 1, 15067 (2015).

    Article  PubMed  Google Scholar 

  146. Coyle, J. T. NMDA receptor and schizophrenia: a brief history. Schizophr. Bull. 38, 920–926 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  147. Fusar-Poli, P. & Meyer-Lindenberg, A. Striatal presynaptic dopamine in schizophrenia, part II: meta-analysis of [18F/11C]-DOPA PET studies. Schizophr. Bull. 39, 33–42 (2013).

    Article  PubMed  Google Scholar 

  148. Fusar-Poli, P. & Meyer-Lindenberg, A. Striatal presynaptic dopamine in schizophrenia, part I: meta-analysis of dopamine active transporter (DAT) density. Schizophr. Bull. 39, 22–32 (2013).

    Article  PubMed  Google Scholar 

  149. Moncrieff, J. A critique of the dopamine hypothesis of schizophrenia and psychosis. Harv. Rev. Psychiatry 17, 214–225 (2009).

    Article  PubMed  Google Scholar 

  150. Susser, E. S. & Lin, S. P. Schizophrenia after prenatal exposure to the Dutch Hunger Winter of 1944–1945. Arch. Gen. Psychiatry 49, 983–988 (1992). This study investigates and reports on schizophrenia incidence following prenatal exposure to the Dutch Hunger Winter; a seminal paper within the epidemiological literature linking early brain development to schizophrenia.

    Article  CAS  PubMed  Google Scholar 

  151. St Clair, D. et al. Rates of adult schizophrenia following prenatal exposure to the Chinese famine of 1959–1961. JAMA 294, 557–562 (2005).

    Article  CAS  PubMed  Google Scholar 

  152. Mackay, E., Dalman, C., Karlsson, H. & Gardner, R. M. Association of gestational weight gain and maternal body mass index in early pregnancy with risk for nonaffective psychosis in offspring. JAMA Psychiatry 74, 339–349 (2017).

    Article  PubMed  Google Scholar 

  153. Brown, A. S. & Derkits, E. J. Prenatal infection and schizophrenia: a review of epidemiologic and translational studies. Am. J. Psychiatry 167, 261–280 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  154. Buka, S. L., Cannon, T. D., Torrey, E. F. & Yolken, R. H. Maternal exposure to herpes simplex virus and risk of psychosis among adult offspring. Biol. Psychiatry 63, 809–815 (2008).

    Article  PubMed  Google Scholar 

  155. Buka, S. L. et al. Maternal infections and subsequent psychosis among offspring. Arch. Gen. Psychiatry 58, 1032–1037 (2001).

    Article  CAS  PubMed  Google Scholar 

  156. Cannon, M., Jones, P. B. & Murray, R. M. Obstetric complications and schizophrenia: historical and meta-analytic review. Am. J. Psychiatry 159, 1080–1092 (2002).

    Article  PubMed  Google Scholar 

  157. Nielsen, P. R., Benros, M. E. & Mortensen, P. B. Hospital contacts with infection and risk of schizophrenia: a population-based cohort study with linkage of Danish national registers. Schizophr. Bull. 40, 1526–1532 (2014).

    Article  PubMed  Google Scholar 

  158. Khandaker, G. M., Zimbron, J., Dalman, C., Lewis, G. & Jones, P. B. Childhood infection and adult schizophrenia: a meta-analysis of population-based studies. Schizophr. Res. 139, 161–168 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  159. Sorensen, H. J. et al. Early developmental milestones and risk of schizophrenia: a 45-year follow-up of the Copenhagen Perinatal Cohort. Schizophr. Res. 118, 41–47 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  160. Hyde, T. M. et al. Enuresis as a premorbid developmental marker of schizophrenia. Brain 131, 2489–2498 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  161. Fish, B., Shapiro, T., Halpern, F. & Wile, R. The prediction of schizophrenia in infancy: a ten-year follow-up report of neurological and psychological development. Am. J. Psychiatry 121, 768–775 (1965).

    Article  CAS  PubMed  Google Scholar 

  162. Isohanni, M. et al. Early developmental milestones in adult schizophrenia and other psychoses. A 31-year follow-up of the Northern Finland 1966 birth cohort. Schizophr. Res. 52, 1–19 (2001).

    Article  CAS  PubMed  Google Scholar 

  163. 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).

    Article  PubMed  PubMed Central  Google Scholar 

  164. Woodberry, K. A., Giuliano, A. J. & Seidman, L. J. Premorbid IQ in schizophrenia: a meta-analytic review. Am. J. Psychiatry 165, 579–587 (2008).

    Article  PubMed  Google Scholar 

  165. Kendler, K. S., Ohlsson, H., Sundquist, J. & Sundquist, K. IQ and schizophrenia in a Swedish national sample: their causal relationship and the interaction of IQ with genetic risk. Am. J. Psychiatry 172, 259–265 (2015).

    Article  PubMed  Google Scholar 

  166. Gottesman, I. I. & Gould, T. D. The endophenotype concept in psychiatry: etymology and strategic intentions. Am. J. Psychiatry 160, 636–645 (2003).

    Article  PubMed  Google Scholar 

  167. Meyer-Lindenberg, A. & Weinberger, D. R. Intermediate phenotypes and genetic mechanisms of psychiatric disorders. Nat. Rev. Neurosci. 7, 818–827 (2006).

    Article  CAS  PubMed  Google Scholar 

  168. Cannon, T. D. et al. Neuropsychological functioning in siblings discordant for schizophrenia and healthy volunteers. Arch. Gen. Psychiatry 51, 651–661 (1994).

    Article  CAS  PubMed  Google Scholar 

  169. Toulopoulou, T. et al. Substantial genetic overlap between neurocognition and schizophrenia: genetic modeling in twin samples. Arch. Gen. Psychiatry 64, 1348–1355 (2007).

    Article  PubMed  Google Scholar 

  170. Birnbaum, R. & Weinberger, D. R. Functional neuroimaging and schizophrenia: a view towards effective connectivity modeling and polygenic risk. Dialogues Clin. Neurosci. 15, 279–289 (2013).

    PubMed  PubMed Central  Google Scholar 

  171. Tan, H. Y., Callicott, J. H. & Weinberger, D. R. Intermediate phenotypes in schizophrenia genetics redux: is it a no brainer? Mol. Psychiatry 13, 233–238 (2008).

    Article  CAS  PubMed  Google Scholar 

  172. Tebbenkamp, A. T., Willsey, A. J., State, M. W. & Sestan, N. The developmental transcriptome of the human brain: implications for neurodevelopmental disorders. Curr. Opin. Neurol. 27, 149–156 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Gallego Romero, I., Pai, A. A., Tung, J. & Gilad, Y. RNA-seq: impact of RNA degradation on transcript quantification. BMC Biol. 12, 42 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Hoffman, G. E. & Schadt, E. E. variancePartition: interpreting drivers of variation in complex gene expression studies. BMC Bioinformatics 17, 483 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  175. Davila, J. I. et al. Impact of RNA degradation on fusion detection by RNA-seq. BMC Genomics 17, 814 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Kleinman, J. E. et al. Genetic neuropathology of schizophrenia: new approaches to an old question, and new uses for postmortem human brains. Biol. Psychiatry 69, 140–145 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Johnson, M. B. et al. Functional and evolutionary insights into human brain development through global transcriptome analysis. Neuron 62, 494–509 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. BrainSeq: A Human Brain Genomics Consortium. BrainSeq: neurogenomics to drive novel target discovery for neuropsychiatric disorders. Neuron 88, 1078–1083 (2015).

    Article  CAS  Google Scholar 

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

  184. Carithers, L. J. et al. A novel approach to high-quality postmortem tissue procurement: the GTEx project. Biopreserv. Biobank. 13, 311–319 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Jaffe, A. E. et al. Developmental regulation of human cortex transcription and its clinical relevance at single base resolution. Nat. Neurosci. 18, 154–161 (2015).

    Article  CAS  PubMed  Google Scholar 

  188. Won, H. et al. Chromosome conformation elucidates regulatory relationships in developing human brain. Nature 538, 523–527 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Jaaro-Peled, H. et al. Neurodevelopmental mechanisms of schizophrenia: understanding disturbed postnatal brain maturation through neuregulin-1-ErbB4 and DISC1. Trends Neurosci. 32, 485–495 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Lambe, E. K., Krimer, L. S. & Goldman-Rakic, P. S. Differential postnatal development of catecholamine and serotonin inputs to identified neurons in prefrontal cortex of rhesus monkey. J. Neurosci. 20, 8780–8787 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Hoftman, G. D. & Lewis, D. A. Postnatal developmental trajectories of neural circuits in the primate prefrontal cortex: identifying sensitive periods for vulnerability to schizophrenia. Schizophr. Bull. 37, 493–503 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors are grateful to A. Jaffe and R. Straub for their constructive comments regarding the manuscript and the data reviewed. The authors are also indebted to T. Hyde and J. Kleinman for their many contributions to the ideas expressed.

Author information

Authors and Affiliations

Authors

Contributions

R.B. and D.R.W. contributed equally to researching data for the article, discussion of content, writing and review and editing of the manuscript before submission.

Corresponding author

Correspondence to Daniel R. Weinberger.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

PowerPoint slides

Glossary

Neural patterning

The process by which cells in the developing nervous system acquire distinct identities according to their specific spatial positions. Controlled by combinatorial action of signalling gradients.

Complex diseases

Diseases caused by a combination of multiple genetic and environmental factors. Examples include cardiovascular disease, type 2 diabetes, autoimmune diseases, cancer, psychiatric disorders and Alzheimer disease.

Single-nucleotide polymorphisms

(SNPs). Variations in a single nucleotide that occurs at a specific position in the genome. The prevalence of any given SNP may be estimated with population-specific reference panels.

Liability scale

The contribution of genetic loci to disease, assuming that individuals have a latent continuous liability of risk of disease that reflects genetic and non-genetic risk factors.

Polygenic risk score

A number based on variation in multiple genetic loci, calculated for a specific individual by summing trait-associated alleles weighted by their estimated effect sizes.

Copy number variants

(CNVs). Structural variations, deletions or duplications of chromosomal segments, typically greater than 1 kb in length, that may affect one or multiple genes, potentially with resulting gene-dosage effects.

Penetrance

The proportion of individuals who harbour a particular variant (or genotype) that express the associated trait or, if a disease-causing mutation, that exhibit clinical symptoms.

Tandem repeat polymorphisms

Variable-number repeat-containing elements or DNA motifs in which one or more nucleotides are repeated directly in adjacency, such as the tri-nucleotide CAG repeats in the gene encoding huntingtin.

Epistatic networks

Networks of non-additive interactions between genotypes at two (or more) loci.

Linkage disequilibrium

(LD). The nonrandom association of alleles at different loci — that is, higher or lower association than if the loci were independent and associated randomly.

Gene set enrichment analyses

An analytic approach to identify classes of genes that are statistically over-represented in a large set of genes and may have an association with a phenotype of interest.

Gene set burden test

An association method of grouping or aggregating variants likely to have similar function instead of testing variants individually.

Expression quantitative trait loci

(eQTLs). Genomic loci or variants that contribute to variation in gene expression levels at proximal or distal loci.

Next-generation RNA sequencing

RNA sequencing that measures the quantity of RNA in a biological sample at a given moment in time, facilitating assessment of gene or transcript differences over time or between groups.

DNA methylation quantitative trait loci

(mQTLs). CpG or other methylation sites in which changes in DNA methylation are associated with a DNA variant at proximal or distal loci.

Induced pluripotent stem cells

(iPSCs). Pluripotent stem cells that are generated by directly reprogramming adult cells and that can be differentiated into specialized cell types while maintaining the genetic background of the individual of origin.

Somatic mosaic events

Within one individual, the presence of two or more cell populations with different genotypes, derived from a post-zygotic mutation, ranging from single-nucleotide to large segmental variations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Birnbaum, R., Weinberger, D. Genetic insights into the neurodevelopmental origins of schizophrenia. Nat Rev Neurosci 18, 727–740 (2017). https://doi.org/10.1038/nrn.2017.125

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrn.2017.125

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