Abstract
Determining the causes of schizophrenia has been a notoriously intractable problem, resistant to a multitude of investigative approaches over centuries. In recent decades, genomic studies have delivered hundreds of robust findings that implicate nearly 300 common genetic variants (via genome-wide association studies) and more than 20 rare variants (via whole-exome sequencing and copy number variant studies) as risk factors for schizophrenia. In parallel, functional genomic and neurobiological studies have provided exceptionally detailed information about the cellular composition of the brain and its interconnections in neurotypical individuals and, increasingly, in those with schizophrenia. Taken together, these results suggest unexpected complexity in the mechanisms that drive schizophrenia, pointing to the involvement of ensembles of genes (polygenicity) rather than single-gene causation. In this Review, we describe what we now know about the genetics of schizophrenia and consider the neurobiological implications of this information.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$29.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$189.00 per year
only $15.75 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout
Similar content being viewed by others
References
American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders 5th edn (American Psychiatric Association, 2013).
World Health Organization. The ICD-10 Classification of Mental and Behavioural Disorders: Diagnostic Criteria for Research (World Health Organization, 1993).
GBD 2019 Mental Disorders Collaborators. Global, regional, and national burden of 12 mental disorders in 204 countries and territories, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet Psychiatry 9, 137–150 (2022).
Knapp, M., Mangalore, R. & Simon, J. The global costs of schizophrenia. Schizophrenia Bull. 30, 279–293 (2004).
Saha, S., Chant, D. & McGrath, J. A systematic review of mortality in schizophrenia: is the differential mortality gap worsening over time? Arch. Gen. Psychiatry 64, 1123–1131 (2007).
Jauhar, S., Johnstone, M. & McKenna, P. J. Schizophrenia. Lancet 399, 473–486 (2022).
McCutcheon, R. A., Reis Marques, T. & Howes, O. D. Schizophrenia — an overview. JAMA Psychiatry 77, 201–210 (2020).
Marder, S. R. & Cannon, T. D. Schizophrenia. N. Engl. J. Med. 381, 1753–1761 (2019).
Goff, D. C. The pharmacologic treatment of schizophrenia-2021. JAMA 325, 175–176 (2021).
Hufner, K., Frajo-Apor, B. & Hofer, A. Neurology issues in schizophrenia. Curr. Psychiatry Rep. 17, 32 (2015).
McGrath, J., Saha, S., Chant, D. & Welham, J. Schizophrenia: a concise overview of incidence, prevalence, and mortality. Epidemiol. Rev. 30, 67–76 (2008).
Kahn, R. S. et al. Schizophrenia. Nat. Rev. Dis. Prim. 1, 15067 (2015).
Johnson, E. C. et al. The relationship between cannabis and schizophrenia: a genetically informed perspective. Addiction 116, 3227–3234 (2021).
Kendler, K. S. The prehistory of psychiatric genetics: 1780–1910. Am. J. Psychiatry 178, 490–508 (2021).
Lichtenstein, P. et al. Common genetic determinants of schizophrenia and bipolar disorder in Swedish families: a population-based study. Lancet 373, 234–239 (2009).
Lichtenstein, P. et al. Recurrence risks for schizophrenia in a Swedish national cohort. Psychol. Med. 36, 1417–1426 (2006).
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).
Wray, N. R. & Gottesman, I. I. Using summary data from the Danish national registers to estimate heritabilities for schizophrenia, bipolar disorder, and major depressive disorder. Front. Genet. 3, 118 (2012).
Sullivan, P. F. & Geschwind, D. H. Defining the genetic, genomic, cellular, and diagnostic architectures of psychiatric disorders. Cell 177, 162–183 (2019).
Karayiorgou, M. et al. Schizophrenia susceptibility associated with interstitial deletions of chromosome 22q11. Proc. Natl Acad. Sci. USA 92, 7612–7616 (1995).
International Schizophrenia Consortium. Common polygenic variation contributes to risk of schizophrenia and bipolar disorder. Nature 460, 748–752 (2009).
Singh, T. et al. Rare loss-of-function variants in SETD1A are associated with schizophrenia and developmental disorders. Nat. Neurosci. 19, 571–577 (2016).
Morris, E., Inglis, A. & Austin, J. Psychiatric genetic counseling for people with copy number variants associated with psychiatric conditions. Clin. Genet. 102, 369–378 (2022).
CNV Working Group of the Psychiatric Genomics Consortium & Schizophrenia Working Group of the Psychiatric Genomics Consortium. Contribution of copy number variants to schizophrenia from a genome-wide study of 41,321 subjects. Nat. Genet. 49, 27–35 (2017).
Singh, T. et al. Rare coding variants in ten genes confer substantial risk for schizophrenia. Nature 604, 509–516 (2022).
Trubetskoy, V. et al. Mapping genomic loci implicates genes and synaptic biology in schizophrenia. Nature 604, 502–508 (2022).
Schizophrenia Working Group of the Psychiatric Genomics Consortium. Biological insights from 108 schizophrenia-associated genetic loci. Nature 511, 421–427 (2014).
Szatkiewicz, J. et al. The genomics of major psychiatric disorders in a large pedigree from Northern Sweden. Transl. Psychiatry 9, 60 (2019).
Fu, J. M. et al. Rare coding variation provides insight into the genetic architecture and phenotypic context of autism. Nat. Genet. 54, 1320–1331 (2022).
Lam, M. et al. Comparative genetic architectures of schizophrenia in East Asian and European populations. Nat. Genet. 51, 1670–1678 (2019).
McClellan, J. & King, M. C. Genomic analysis of mental illness: a changing landscape. JAMA 303, 2523–2524 (2010).
Andrade-Guerrero, J. et al. Alzheimer’s disease: an updated overview of its genetics. Int. J. Mol. Sci. 24, 3754 (2023).
Halvorsen, M. et al. Increased burden of ultra-rare structural variants localizing to boundaries of topologically associated domains in schizophrenia. Nat. Commun. 11, 1842 (2020).
Murray, G. K. et al. Could polygenic risk scores be useful in psychiatry?: a review. JAMA Psychiatry 78, 210–219 (2021).
Visscher, P. M., Yengo, L., Cox, N. J. & Wray, N. R. Discovery and implications of polygenicity of common diseases. Science 373, 1468–1473 (2021).
Wray, N. R. et al. From basic science to clinical application of polygenic risk scores: a primer. JAMA Psychiatry 78, 101–109 (2021).
Sullivan, P. F. et al. Leveraging base-pair mammalian constraint to understand genetic variation and human disease. Science 380, eabn2937 (2023).
Khera, A. V. et al. Genome-wide polygenic scores for common diseases identify individuals with risk equivalent to monogenic mutations. Nat. Genet. 50, 1219–1224 (2018).
Farrell, M. S. et al. Evaluating historical candidate genes for schizophrenia. Mol. Psychiatry 20, 555–562 (2015).
Franke, B. et al. Genetic influences on schizophrenia and subcortical brain volumes: large-scale proof of concept. Nat. Neurosci. 19, 420–431 (2016).
van Erp, T. G. et al. Subcortical brain volume abnormalities in 2028 individuals with schizophrenia and 2540 healthy controls via the ENIGMA consortium. Mol. Psychiatry 21, 547–553 (2016).
Grasby, K. L. et al. The genetic architecture of the human cerebral cortex. Science 367, eaay6690 (2020).
Gutman, B. A. et al. A meta-analysis of deep brain structural shape and asymmetry abnormalities in 2,833 individuals with schizophrenia compared with 3,929 healthy volunteers via the ENIGMA Consortium. Hum. Brain Mapp. 43, 352–372 (2022).
Schijven, D. et al. Large-scale analysis of structural brain asymmetries in schizophrenia via the ENIGMA consortium. Proc. Natl Acad. Sci. USA 120, e2213880120 (2023).
Boyle, E. A., Li, Y. I. & Pritchard, J. K. An expanded view of complex traits: from polygenic to omnigenic. Cell 169, 1177–1186 (2017).
Wray, N. R., Wijmenga, C., Sullivan, P. F., Yang, J. & Visscher, P. M. Common disease is more complex than implied by the core gene omnigenic model. Cell 173, 1573–1580 (2018).
Wainschtein, P. et al. Assessing the contribution of rare variants to complex trait heritability from whole-genome sequence data. Nat. Genet. 54, 263–273 (2022).
Pingault, J. B. et al. Using genetic data to strengthen causal inference in observational research. Nat. Rev. Genet. 19, 566–580 (2018).
Saccaro, L. F., Gasparini, S. & Rutigliano, G. Applications of Mendelian randomization in psychiatry: a comprehensive systematic review. Psychiatr. Genet. 32, 199–213 (2022).
Said, S. et al. Genetic analysis of over half a million people characterises C-reactive protein loci. Nat. Commun. 13, 2198 (2022).
Revez, J. A. et al. Genome-wide association study identifies 143 loci associated with 25 hydroxyvitamin D concentration. Nat. Commun. 11, 1647 (2020).
Christmas, M. J. et al. Evolutionary constraint and innovation across hundreds of placental mammals. Science 380, eabn3943 (2023).
Giniatullina, A. et al. Functional characterization of the PCLO p.Ser4814Ala variant associated with major depressive disorder reveals cellular but not behavioral differences. Neuroscience 300, 518–538 (2015).
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).
Rummel, C. K. et al. Massively parallel functional dissection of schizophrenia-associated noncoding genetic variants. Cell 186, 5165–5182 (2023).
Li, Y. E. et al. A comparative atlas of single-cell chromatin accessibility in the human brain. Science 382, eadf7044 (2023).
Dekker, J. Mapping the 3D genome: aiming for consilience. Nat. Rev. Mol. Cell Biol. 17, 741–742 (2016).
Won, H. et al. Chromosome conformation elucidates regulatory relationships in developing human brain. Nature 538, 523–527 (2016).
Tan, L. et al. Lifelong restructuring of 3D genome architecture in cerebellar granule cells. Science 381, 1112–1119 (2023).
Fiziev, P. & Ernst, J. ChromTime: modeling spatio-temporal dynamics of chromatin marks. Genome Biol. 19, 109 (2018).
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).
Howes, O. D. & Kapur, S. The dopamine hypothesis of schizophrenia: version III — the final common pathway. Schizophr. Bull. 35, 549–562 (2009).
Howes, O. D. & Onwordi, E. C. The synaptic hypothesis of schizophrenia version III: a master mechanism. Mol. Psychiatry 28, 1843–1856 (2023).
Osimo, E. F., Beck, K., Reis Marques, T. & Howes, O. D. Synaptic loss in schizophrenia: a meta-analysis and systematic review of synaptic protein and mRNA measures. Mol. Psychiatry 24, 549–561 (2019).
Trepanier, M. O., Hopperton, K. E., Mizrahi, R., Mechawar, N. & Bazinet, R. P. Postmortem evidence of cerebral inflammation in schizophrenia: a systematic review. Mol. Psychiatry 21, 1009–1026 (2016).
Weinstein, J. J. et al. Pathway-specific dopamine abnormalities in schizophrenia. Biol. Psychiatry 81, 31–42 (2017).
Davalieva, K., Maleva Kostovska, I. & Dwork, A. J. Proteomics research in schizophrenia. Front. Cell Neurosci. 10, 18 (2016).
Laskaris, L. E. et al. Microglial activation and progressive brain changes in schizophrenia. Br. J. Pharmacol. 173, 666–680 (2016).
Schwerk, A., Alves, F. D., Pouwels, P. J. & van Amelsvoort, T. Metabolic alterations associated with schizophrenia: a critical evaluation of proton magnetic resonance spectroscopy studies. J. Neurochem. 128, 1–87 (2014).
Dean, B. Neurochemistry of schizophrenia: the contribution of neuroimaging postmortem pathology and neurochemistry in schizophrenia. Curr. Top. Med. Chem. 12, 2375–2392 (2012).
Gonzalez-Burgos, G. & Lewis, D. A. NMDA receptor hypofunction, parvalbumin-positive neurons, and cortical gamma oscillations in schizophrenia. Schizophr. Bull. 38, 950–957 (2012).
Sohal, V. S. Neurobiology of schizophrenia. Curr. Opin. Neurobiol. 84, 102820 (2023).
Glantz, L. A. & Lewis, D. A. Decreased dendritic spine density on prefrontal cortical pyramidal neurons in schizophrenia. Arch. Gen. Psychiatry 57, 65–73 (2000).
Dienel, S. J., Fish, K. N. & Lewis, D. A. The nature of prefrontal cortical GABA neuron alterations in schizophrenia: markedly lower somatostatin and parvalbumin gene expression without missing neurons. Am. J. Psychiatry 180, 495–507 (2023).
Aryal, S. et al. Deep proteomics identifies shared molecular pathway alterations in synapses of patients with schizophrenia and bipolar disorder and mouse model. Cell Rep. 42, 112497 (2023).
Walker, R. L. et al. Genetic control of expression and splicing in developing human brain informs disease mechanisms. Cell 179, 750–771 (2019).
Benjamin, K. J. M. et al. Analysis of the caudate nucleus transcriptome in individuals with schizophrenia highlights effects of antipsychotics and new risk genes. Nat. Neurosci. 25, 1559–1568 (2022).
Kim, M. et al. Brain gene co-expression networks link complement signaling with convergent synaptic pathology in schizophrenia. Nat. Neurosci. 24, 799–809 (2021).
Batiuk, M. Y. et al. Upper cortical layer-driven network impairment in schizophrenia. Sci. Adv. 8, eabn8367 (2022).
Ruzicka, W. et al. Single-cell multi-cohort dissection of the schizophrenia transcriptome. Science 384, eadg5136 (2024).
Aguilar, D. D. & McNally, J. M. Subcortical control of the default mode network: role of the basal forebrain and implications for neuropsychiatric disorders. Brain Res. Bull. 185, 129–139 (2022).
Medoff, D. R., Holcomb, H. H., Lahti, A. C. & Tamminga, C. A. Probing the human hippocampus using rCBF: contrasts in schizophrenia. Hippocampus 11, 543–550 (2001).
Perrottelli, A., Giordano, G. M., Brando, F., Giuliani, L. & Mucci, A. EEG-based measures in at-risk mental state and early stages of schizophrenia: a systematic review. Front. Psychiatry 12, 653642 (2021).
Amann, L. C. et al. Mouse behavioral endophenotypes for schizophrenia. Brain Res. Bull. 83, 147–161 (2010).
Birnbaum, R. & Weinberger, D. R. Genetic insights into the neurodevelopmental origins of schizophrenia. Nat. Rev. Neurosci. 18, 727–740 (2017).
Murray, R. M. & Lewis, S. W. Is schizophrenia a neurodevelopmental disorder? Br. Med. J. 295, 681–682 (1987).
Weinberger, D. R. Implications of normal brain development for the pathogenesis of schizophrenia. Arch. Gen. Psychiatry 44, 660–669 (1987).
Owen, M. J., O’Donovan, M. C., Thapar, A. & Craddock, N. Neurodevelopmental hypothesis of schizophrenia. Br. J. Psychiatry 198, 173–175 (2011).
Mortensen, P. B. et al. Effects of family history and place and season of birth on the risk of schizophrenia. N. Engl. J. Med. 340, 603–608 (1999).
Bryois, J. et al. Genetic identification of cell types underlying brain complex traits yields insights into the etiology of Parkinson’s disease. Nat. Genet. 52, 482–493 (2020).
Yao, S. et al. Connecting genomic results for psychiatric disorders to human brain 1 cell types and regions reveals convergence with functional connectivity. Preprint at medRxiv https://doi.org/10.1101/2024.01.18.24301478 (2024).
Thyme, S. B. et al. Phenotypic landscape of schizophrenia-associated genes defines candidates and their shared functions. Cell 177, 478–491 (2019).
Sekar, A. et al. Schizophrenia risk from complex variation of complement component 4. Nature 530, 177–183 (2016).
Hyman, S. E. & Nestler, E. J. Initiation and adaptation: a paradigm for understanding psychotropic drug action. Am. J. Psychiatry 153, 151–162 (1996).
Hyman, S. E. Use of mouse models to investigate the contributions of CNVs associated with schizophrenia and autism to disease mechanisms. Curr. Opin. Genet. Dev. 68, 99–105 (2021).
Zhang, W. et al. Mouse genome rewriting and tailoring of three important disease loci. Nature 623, 423–431 (2023).
Jin, X. et al. In vivo perturb-seq reveals neuronal and glial abnormalities associated with autism risk genes. Science 370, eaaz6063 (2020).
Zhang, S. et al. Multiple genes in a single GWAS risk locus synergistically mediate aberrant synaptic development and function in human neurons. Cell Genom. 3, 100399 (2023).
Schrode, N. et al. Synergistic effects of common schizophrenia risk variants. Nat. Genet. 51, 1475–1485 (2019).
Page, S. C. et al. Electrophysiological measures from human iPSC-derived neurons are associated with schizophrenia clinical status and predict individual cognitive performance. Proc. Natl Acad. Sci. USA 119, e2109395119 (2022).
Stachowiak, E. K. et al. Cerebral organoids reveal early cortical maldevelopment in schizophrenia-computational anatomy and genomics, role of FGFR1. Transl. Psychiatry 7, 6 (2017).
Brennand, K. et al. Phenotypic differences in hiPSC NPCs derived from patients with schizophrenia. Mol. Psychiatry 20, 361–368 (2015).
Brennand, K. J. et al. Modelling schizophrenia using human induced pluripotent stem cells. Nature 473, 221–225 (2011).
Mollon, J., Almasy, L., Jacquemont, S. & Glahn, D. C. The contribution of copy number variants to psychiatric symptoms and cognitive ability. Mol. Psychiatry 28, 1480–1493 (2023).
Mulle, J. G., Sullivan, P. F. & Hjerling-Leffler, J. Editorial overview: rare CNV disorders and neuropsychiatric phenotypes: opportunities, challenges, solutions. Curr. Opin. Genet. Dev. 68, iii–ix (2021).
Marshall, 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).
Fung, W. L. et al. Practical guidelines for managing adults with 22q11.2 deletion syndrome. Genet. Med. 17, 599–609 (2015).
Moreno-De-Luca, D. & Martin, C. L. All for one and one for all: heterogeneity of genetic etiologies in neurodevelopmental psychiatric disorders. Curr. Opin. Genet. Dev. 68, 71–78 (2021).
Tansey, K. E. et al. Common alleles contribute to schizophrenia in CNV carriers. Mol. Psychiatry 21, 1085–1089 (2016).
Silva, A. I. et al. Neuroimaging findings in neurodevelopmental copy number variants: identifying molecular pathways to convergent phenotypes. Biol. Psychiatry 92, 341–361 (2022).
Caseras, X. et al. Effects of genomic copy number variants penetrant for schizophrenia on cortical thickness and surface area in healthy individuals: analysis of the UK Biobank. Br. J. Psychiatry 218, 104–111 (2021).
Gordon, A. et al. Transcriptomic networks implicate neuronal energetic abnormalities in three mouse models harboring autism and schizophrenia-associated mutations. Mol. Psychiatry 26, 1520–1534 (2021).
Sebastian, R. et al. Schizophrenia-associated NRXN1 deletions induce developmental-timing- and cell-type-specific vulnerabilities in human brain organoids. Nat. Commun. 14, 3770 (2023).
Nehme, R. et al. The 22q11.2 region regulates presynaptic gene-products linked to schizophrenia. Nat. Commun. 13, 3690 (2022).
Pak, C. et al. Cross-platform validation of neurotransmitter release impairments in schizophrenia patient-derived NRXN1-mutant neurons. Proc. Natl Acad. Sci. USA 118, e2025598118 (2021).
Khan, T. A. et al. Neuronal defects in a human cellular model of 22q11.2 deletion syndrome. Nat. Med. 26, 1888–1898 (2020).
Flaherty, E. et al. Neuronal impact of patient-specific aberrant NRXN1alpha splicing. Nat. Genet. 51, 1679–1690 (2019).
Parnell, E. et al. Excitatory dysfunction drives network and calcium handling deficits in 16p11.2 duplication schizophrenia induced pluripotent stem cell-derived neurons. Biol. Psychiatry 94, 153–163 (2023).
Farsi, Z. et al. Brain-region-specific changes in neurons and glia and dysregulation of dopamine signaling in Grin2a mutant mice. Neuron 111, 3378–3396 (2023).
Mukai, J. et al. Recapitulation and reversal of schizophrenia-related phenotypes in setd1a-deficient mice. Neuron 104, 471–487 e412 (2019).
Fromer, M. et al. De novo mutations in schizophrenia implicate synaptic networks. Nature 506, 179–184 (2014).
Genovese, G. et al. Increased burden of ultra-rare protein-altering variants among 4,877 individuals with schizophrenia. Nat. Neurosci. 19, 1433–1441 (2016).
Koopmans, F. et al. SynGO: an evidence-based, expert-curated knowledge base for the synapse. Neuron 103, 217–234 (2019).
Finucane, H. K. et al. Heritability enrichment of specifically expressed genes identifies disease-relevant tissues and cell types. Nat. Genet. 50, 621–629 (2018).
Ongen, H. et al. Estimating the causal tissues for complex traits and diseases. Nat. Genet. 49, 1676–1683 (2017).
Li, M. et al. Integrative functional genomic analysis of human brain development and neuropsychiatric risks. Science 362, eaat7615 (2018).
Tian, W. et al. Single-cell DNA methylation and 3D genome architecture in the human brain. Science 382, eadf5357 (2023).
Zhu, K. et al. Multi-omic profiling of the developing human cerebral cortex at the single-cell level. Sci. Adv. 9, eadg3754 (2023).
Jagadeesh, K. A. et al. Identifying disease-critical cell types and cellular processes by integrating single-cell RNA-sequencing and human genetics. Nat. Genet. 54, 1479–1492 (2022).
Zhang, M. J. et al. Polygenic enrichment distinguishes disease associations of individual cells in single-cell RNA-seq data. Nat. Genet. 54, 1572–1580 (2022).
Wang, R., Lin, D. Y. & Jiang, Y. EPIC: inferring relevant cell types for complex traits by integrating genome-wide association studies and single-cell RNA sequencing. PLoS Genet. 18, e1010251 (2022).
Ziffra, R. S. et al. Single-cell epigenomics reveals mechanisms of human cortical development. Nature 598, 205–213 (2021).
Watanabe, K., Umicevic Mirkov, M., de Leeuw, C. A., van den Heuvel, M. P. & Posthuma, D. Genetic mapping of cell type specificity for complex traits. Nat. Commun. 10, 3222 (2019).
Skene, N. G. et al. Genetic identification of brain cell types underlying schizophrenia. Nat. Genet. 50, 825–833 (2018).
Lake, B. B. et al. Integrative single-cell analysis of transcriptional and epigenetic states in the human adult brain. Nat. Biotechnol. 36, 70–80 (2018).
Calderon, D. et al. Inferring relevant cell types for complex traits by using single-cell gene expression. Am. J. Hum. Genet. 101, 686–699 (2017).
Siletti, K. et al. Transcriptomic diversity of cell types across the adult human brain. Science 382, eadd7046 (2023).
Munguba, H. et al. Postnatal Sox6 regulates synaptic function of cortical parvalbumin-expressing neurons. J. Neurosci. 41, 8876–8886 (2021).
Leek, J. T. et al. Tackling the widespread and critical impact of batch effects in high-throughput data. Nat. Rev. Genet. 11, 733–739 (2010).
Strittmatter, W. J. et al. Apolipoprotein E: high-avidity binding to beta-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer disease. Proc. Natl Acad. Sci. USA 90, 1977–1981 (1993).
Horvath, G. A., Stowe, R. M., Ferreira, C. R. & Blau, N. Clinical and biochemical footprints of inherited metabolic diseases. III. Psychiatric presentations. Mol. Genet. Metab. 130, 1–6 (2020).
Taliun, D. et al. Sequencing of 53,831 diverse genomes from the NHLBI TOPMed Program. Nature 590, 290–299 (2021).
Mukamel, R. E. et al. Repeat polymorphisms underlie top genetic risk loci for glaucoma and colorectal cancer. Cell 186, 3659–3673.e23 (2023).
Hanks, S. C. et al. Extent to which array genotyping and imputation with large reference panels approximate deep whole-genome sequencing. Am. J. Hum. Genet. 109, 1653–1666 (2022).
Peterson, R. E. et al. Genome-wide association studies in ancestrally diverse populations: opportunities, methods, pitfalls, and recommendations. Cell 179, 589–603 (2019).
Graham, S. E. et al. The power of genetic diversity in genome-wide association studies of lipids. Nature 600, 675–679 (2021).
Zhu, Z. et al. Integration of summary data from GWAS and eQTL studies predicts complex trait gene targets. Nat. Genet. 48, 481–487 (2016).
Wolter, J. M. et al. Cellular genome-wide association study identifies common genetic variation influencing lithium-induced neural progenitor proliferation. Biol. Psychiatry 93, 8–17 (2023).
GTEx Consortium. The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science 369, 1318–1330 (2020).
Zhang, S. et al. Allele-specific open chromatin in human iPSC neurons elucidates functional disease variants. Science 369, 561–565 (2020).
Forrest, M. P. et al. Open chromatin profiling in hiPSC-derived neurons prioritizes functional noncoding psychiatric risk variants and highlights neurodevelopmental loci. Cell Stem Cell 21, 305–318.e8 (2017).
Encode Project Consortium et al. Perspectives on ENCODE. Nature 583, 693–698 (2020).
Hawrylycz, M. et al. A guide to the BRAIN initiative cell census network data ecosystem. PLoS Biol. 21, e3002133 (2023).
Sey, N. Y. A., Pratt, B. M. & Won, H. Annotating genetic variants to target genes using H-MAGMA. Nat. Protoc. 18, 22–35 (2023).
Rajarajan, P. et al. Neuron-specific signatures in the chromosomal connectome associated with schizophrenia risk. Science 362, eaat4311 (2018).
McAfee, J. C. et al. Systematic investigation of allelic regulatory activity of schizophrenia-associated common variants. Cell Genom. 3, 100404 (2023).
Klann, T. S. et al. CRISPR–Cas9 epigenome editing enables high-throughput screening for functional regulatory elements in the human genome. Nat. Biotechnol. 35, 561–568 (2017).
Yang, X. et al. Functional characterization of gene regulatory elements and neuropsychiatric disease-associated risk loci in iPSCs and iPSC-derived neurons. Preprint at bioRxiv https://doi.org/10.1101/2023.08.30.555359 (2023).
Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576, 149–157 (2019).
Komor, A. C., Kim, Y. B., Packer, M. S., Zuris, J. A. & Liu, D. R. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533, 420–424 (2016).
Ren, X. et al. High throughput PRIME editing screens identify functional DNA variants in the human genome. Mol. Cell 83, 4633–4645 (2023).
Acknowledgements
J.H.-L. was supported by the Swedish Research Council (Vetenskapsrådet, award 2018-00799), Swedish Brain Foundation (Hjärnfonden, award FO2018-0272) and European Research Council (SCHIZTYPE, grant agreement 819540). P.F.S. was supported by the Swedish Research Council (Vetenskapsrådet, award D0886501) and the US National Institute of Mental Health (R01s MH124871, MH121545 and MH123724).
Author information
Authors and Affiliations
Contributions
All authors researched data for the article and contributed substantially to discussion of the content. P.F.S. and J.H-L. wrote the article and reviewed and/or edited the manuscript before submission.
Corresponding authors
Ethics declarations
Competing interests
P.F.S. is a consultant and shareholder for Neumora Therapeutics.
Peer review
Peer review information
Nature Reviews Neuroscience thanks Kristen Brennand, Brien Riley and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Related links
Psychiatric Genetics Consortium: https://pgc.unc.edu/
Supplementary information
Glossary
- Brain organoids
-
Three-dimensional cell cultures that mimic the structure and function of the brain, derived from pluripotent stem cells.
- Copy number variants
-
(CNVs). Structural variations in the genome, in which large sections of DNA (containing from one gene to many genes) are duplicated or deleted, potentially influencing traits or diseases.
- Dopamine hypothesis
-
A theory proposing that dysregulation of dopamine neurotransmission in the brain contributes to the development of schizophrenia.
- Epigenomic
-
Referring to chemical modifications to DNA and histone proteins that regulate gene expression without altering the DNA sequence.
- Genetic risk factors
-
Variations in the DNA sequence that increase the likelihood of developing a particular trait or disease (such as schizophrenia).
- Genetic variants
-
Measurable differences in DNA sequence among individuals, including SNPs, coding and non-coding variants, copy number variants, insertion and deletions.
- Genome-wide association study
-
(GWAS). A method to identify genetic variations across the entire genome associated with traits or diseases.
- Heritability
-
The proportion of the total variation in a trait in a population that is owing to genetic differences among individuals.
- Induced pluripotent stem cell
-
An adult cell reprogrammed to exhibit embryonic stem-cell-like properties and capable of differentiating into various cell types.
- Linkage disequilibrium
-
The nonrandom association or correlation of alleles at different loci within a population. Linkage disequilibrium is detectible between pairs of genetic markers with tens of kilobases but may span many megabases in specific regions.
- Mendelian randomization
-
A method using genetic variants as instrumental variables to investigate causal relationships between modifiable exposures and health outcomes.
- Polygenic risk score
-
(PRS). A numerical score calculated from multiple genetic variants associated with a trait or disease, used as a summation of genetic predisposition of an individual.
- Single-cell RNA sequencing
-
A sequencing technique to analyse gene expression in single cells (or single nuclei), providing insights into cellular heterogeneity and functional diversity.
- SNPs
-
Variations in a single nucleotide at a specific position in the genome.
- Whole-exome sequencing
-
(WES). Sequencing of the protein-coding regions of the genome.
- Whole-genome sequencing
-
Sequencing of the entire genome, including coding and non-coding regions, providing a comprehensive view of the genetic makeup of an individual.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Sullivan, P.F., Yao, S. & Hjerling-Leffler, J. Schizophrenia genomics: genetic complexity and functional insights. Nat. Rev. Neurosci. 25, 611–624 (2024). https://doi.org/10.1038/s41583-024-00837-7
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s41583-024-00837-7