Skip to main content

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

  • Article
  • Published:

Depression pathophysiology, risk prediction of recurrence and comorbid psychiatric disorders using genome-wide analyses

Abstract

Depression is a common psychiatric disorder and a leading cause of disability worldwide. Here we conducted a genome-wide association study meta-analysis of six datasets, including >1.3 million individuals (371,184 with depression) and identified 243 risk loci. Overall, 64 loci were new, including genes encoding glutamate and GABA receptors, which are targets for antidepressant drugs. Intersection with functional genomics data prioritized likely causal genes and revealed new enrichment of prenatal GABAergic neurons, astrocytes and oligodendrocyte lineages. We found depression to be highly polygenic, with ~11,700 variants explaining 90% of the single-nucleotide polymorphism heritability, estimating that >95% of risk variants for other psychiatric disorders (anxiety, schizophrenia, bipolar disorder and attention deficit hyperactivity disorder) were influencing depression risk when both concordant and discordant variants were considered, and nearly all depression risk variants influenced educational attainment. Additionally, depression genetic risk was associated with impaired complex cognition domains. We dissected the genetic and clinical heterogeneity, revealing distinct polygenic architectures across subgroups of depression and demonstrating significantly increased absolute risks for recurrence and psychiatric comorbidity among cases of depression with the highest polygenic burden, with considerable sex differences. The risks were up to 5- and 32-fold higher than cases with the lowest polygenic burden and the background population, respectively. These results deepen the understanding of the biology underlying depression, its disease progression and inform precision medicine approaches to treatment.

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

Access options

Buy this article

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

Fig. 1: Manhattan plots of GWAS, gene-based analysis and TWAS of DEP.
Fig. 2: Genetic overlap between DEP and selected phenotypes using MiXeR.
Fig. 3: Cell-type enrichments.
Fig. 4: Association of DEP PRS with cognitive abilities.
Fig. 5: Absolute risks of recurrency and developing comorbid disorders stratified by polygenic scores.

Similar content being viewed by others

Data availability

Supplementary Figs. 1–15 and 24–28 are available at https://doi.org/10.6084/m9.figshare.22139849. The GWAS meta-analysis summary statistics from this publication, not including 23andMe, are available at https://ipsych.dk/en/research/downloads/. To access the summary statistics from the meta-analysis of all cohorts, including 23andMe, a data transfer agreement is required from 23andMe (dataset-request@23andMe.com) before a request is made to the corresponding authors. See https://research.23andme.com/collaborate/#dataset-access/ for more information and to apply for access to the data.

All relevant iPSYCH data are available from the authors after approval by the iPSYCH Data Access Committee and can only be accessed on the secured Danish server (GenomeDK, https://genome.au.dk) as the data are protected by Danish legislation. For data access please contact A.D.B.

The downloadable data of the HRC were used for imputation: http://www.haplotype-reference-consortium.org/.

Data used for brain transcriptome model generation are available from PsychENCODE (http://resource.psychencode.org/); genotypes are controlled data and access instructions are provided at https://www.synapse.org/#!Synapse:syn4921369/wiki/477467.

Resources for the colocalization analysis are available at PsychENCODE (http://resource.psychencode.org/) and ROSMAP.

Note that some datasets have been indirectly accessed at the FUMA website. In general, PsychENCODE (http://resource.psychencode.org/) was used for SNP annotations (enhancer, H3K27ac markers), eQTLs and HiC-based enhancer–promoter interactions. GTEx v.6/v.7/v.8 eQTLs and gene expression used in the pipeline were obtained from GTEx (http://www.gtexportal.org/home/). The following eQTL datasets in FUMA were used for gene mapping: BrainSeq (http://eqtl.brainseq.org/), PsychENCODE eQTLs (http://resource.psychencode.org/), Common Mind Consortium (https://www.synapse.org//#!Synapse:syn5585484), BRAINEAC (http://www.braineac.org/) and GTEx/V8/Brain (https://www.gtexportal.org/home/datasets/). Chromatin interaction datasets in FUMA used for gene mapping were PsychENCODE eQTLs and HiC-based enhancer–promoter interactions (http://resource.psychencode.org/), HiC (https://doi.org/10.1101/406330, GSE87112) and Roadmap–brain (https://egg2.wustl.edu/roadmap/web_portal/DNase_reg.html). The following single-cell RNA-seq datasets were used in the cell-type specific analyses in FUMA (https://fuma.ctglab.nl/tutorial#datasets): PsychENCODE human developmental and adult brain samples (http://resource.psychencode.org/), Allen Brain Atlas Cell Type (http://celltypes.brain-map.org/api/v2/well_known_file_download/694416667), DroNc human brain samples (hippocampus) (https://portals.broadinstitute.org/single_cell#study-dronc-seq-single-nucleus-rna-seq-on-human-archived-brain, https://www.gtexportal.org/), human prefrontal cortex brain samples (GSE104276), human cortex brain samples (GSE67835), Linarsson human temporal cortex brain samples (GSE101601) and human midbrain samples (GSE76381).

GO of Biological Process and Cellular Components datasets of MSigDB v.7.0 (https://www.gsea-msigdb.org/gsea/msigdb) were used for the gene-set enrichment analysis in FUMA’s GENE2FUNC module. Please refer to https://fuma.ctglab.nl/links and https://fuma.ctglab.nl/tutorial#datasets for additional information on availability of datasets.

Code availability

Software and analytical methods used in data analyses include:

Ricopili: https://sites.google.com/a/broadinstitute.org/ricopili/.

Eigensoft v.6.1.4: https://www.hsph.harvard.edu/alkes-price/software/ and https://github.com/chrchang/eigensoft.

Eagel v.2.3.5: https://github.com/poruloh/Eagle.

Minimac3: https://github.com/Santy-8128/Minimac3.

METAL v.2011-03-05: https://genome.sph.umich.edu/wiki/METAL_Documentation.

Genotyping and imputation with the Finnish population-specific SISu v.3 reference panel: https://www.protocols.io/view/genotype-imputation-workflow-v3-0-xbgfijw.

SAIGE v.0.20: Scalable and Accurate Implementation of GEneralized mixed model v.0.20: https://github.com/weizhouUMICH/SAIGE/.

LDsc v.1.0.1: https://github.com/bulik/ldsc.

MiXeR v.1.3: https://github.com/precimed/mixer.

LAVA 2022-09-29 https://ctg.cncr.nl/software/lava and https://github.com/josefin-werme/LAVA. https://github.com/josefin-werme/LAVA. The method and code used for genome partitioning used for the LAVA analyses is available at https://github.com/cadeleeuw/lava-partitioning.

GCTA v.1.93.2: https://yanglab.westlake.edu.cn/software/gcta/#Overview.

LDpred2: https://privefl.github.io/bigsnpr/articles/LDpred2.html.

FUMA v.1.3.7: https://fuma.ctglab.nl/.

S-PrediXcan: https://github.com/hakyimlab/MetaXcan.

eCAVIAR: http://zarlab.cs.ucla.edu/tag/ecaviar/.

coloc v.2: https://github.com/Stahl-Lab-MSSM/coloc2.

CAUSALdb finemapping pipeline: https://github.com/mulinlab/CAUSALdb-finemapping-pip.

Variance partition fraction for GLM with linear, logistic or probit regression used for PNC analysis: https://bioconductor.org/packages/variancePartition

Cox proportional hazard modeling: the R packages survival v.3.4-0 (https://CRAN.R-project.org/package=survival).

R v.4.2.2 was used in general for statistical analyses and plotting (https://www.Rproject.org).

References

  1. Kessler, R. C. & Bromet, E. J. The epidemiology of depression across cultures. Annu. Rev. Public Health 34, 119–138 (2013).

    PubMed  PubMed Central  Google Scholar 

  2. Hasin, D. S. et al. Epidemiology of adult DSM-5 major depressive disorder and its specifiers in the United States. JAMA Psychiatry 75, 336–346 (2018).

    PubMed  PubMed Central  Google Scholar 

  3. Pedersen, C. B. et al. A comprehensive nationwide study of the incidence rate and lifetime risk for treated mental disorders. JAMA Psychiatry 71, 573–581 (2014).

    PubMed  Google Scholar 

  4. Lopez, A. D., Mathers, C. D., Ezzati, M., Jamison, D. T. & Murray, C. J. Global and regional burden of disease and risk factors, 2001: systematic analysis of population health data. Lancet 367, 1747–1757 (2006).

    PubMed  Google Scholar 

  5. Wittchen, H. U. et al. The size and burden of mental disorders and other disorders of the brain in Europe 2010. Eur. Neuropsychopharmacol. 21, 655–679 (2011).

    CAS  PubMed  Google Scholar 

  6. Gustavsson, A. et al. Cost of disorders of the brain in Europe 2010. Eur. Neuropsychopharmacol. 21, 718–779 (2011).

    CAS  PubMed  Google Scholar 

  7. Angst, F., Stassen, H. H., Clayton, P. J. & Angst, J. Mortality of patients with mood disorders: follow-up over 34-38 years. J. Affect. Disord. 68, 167–181 (2002).

    CAS  PubMed  Google Scholar 

  8. Ferrari, A. J. et al. Burden of depressive disorders by country, sex, age, and year: findings from the global burden of disease study 2010. PLoS Med. 10, e1001547 (2013).

    PubMed  PubMed Central  Google Scholar 

  9. Plana-Ripoll, O. et al. Exploring comorbidity within mental disorders among a Danish national population. JAMA Psychiatry 76, 259–270 (2019).

    PubMed  PubMed Central  Google Scholar 

  10. McGrath, J. J. et al. Comorbidity within mental disorders: a comprehensive analysis based on 145990 survey respondents from 27 countries. Epidemiol. Psychiatr. Sci. 29, e153 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Sullivan, P. F., Neale, M. C. & Kendler, K. S. Genetic epidemiology of major depression: review and meta-analysis. Am. J. Psychiatry 157, 1552–1562 (2000).

    CAS  PubMed  Google Scholar 

  12. Wray, N. R. et al. Genome-wide association analyses identify 44 risk variants and refine the genetic architecture of major depression. Nat. Genet. 50, 668–681 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Demontis, D. et al. Discovery of the first genome-wide significant risk loci for attention deficit/hyperactivity disorder. Nat. Genet. 51, 63–75 (2018).

    PubMed  PubMed Central  Google Scholar 

  14. Howard, D. M. et al. Genome-wide meta-analysis of depression identifies 102 independent variants and highlights the importance of the prefrontal brain regions. Nat. Neurosci. 22, 343–352 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Grove, J. et al. Identification of common genetic risk variants for autism spectrum disorder. Nat. Genet. 51, 431–444 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Quello, S. B., Brady, K. T. & Sonne, S. C. Mood disorders and substance use disorder: a complex comorbidity. Sci. Pr. Perspect. 3, 13–21 (2005).

    Google Scholar 

  17. Davis, L., Uezato, A., Newell, J. M. & Frazier, E. Major depression and comorbid substance use disorders. Curr. Opin. Psychiatry 21, 14–18 (2008).

    PubMed  Google Scholar 

  18. Murray, G. K. et al. Could polygenic risk scores be useful in psychiatry?: a review. JAMA Psychiatry 78, 210–219 (2021).

    PubMed  Google Scholar 

  19. Rees, E. & Owen, M. J. Translating insights from neuropsychiatric genetics and genomics for precision psychiatry. Genome Med. 12, 43 (2020).

    PubMed  PubMed Central  Google Scholar 

  20. Perkins, D. O. et al. Polygenic risk score contribution to psychosis prediction in a target population of persons at clinical high risk. Am. J. Psychiatry 177, 155–163 (2020).

    PubMed  Google Scholar 

  21. Howard, D. M. et al. Genome-wide association study of depression phenotypes in UK Biobank identifies variants in excitatory synaptic pathways. Nat. Commun. 9, 1470 (2018).

    PubMed  PubMed Central  Google Scholar 

  22. Kurki, M. I. et al. FinnGen provides genetic insights from a well-phenotyped isolated population. Nature 613, 508–518 (2023).

  23. Hyde, C. L. et al. Identification of 15 genetic loci associated with risk of major depression in individuals of European descent. Nat. Genet. 48, 1031–1036 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Levey, D. F. et al. Bi-ancestral depression GWAS in the Million Veteran Program and meta-analysis in >1.2 million individuals highlight new therapeutic directions. Nat. Neurosci. 24, 954–963 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Bybjerg-Grauholm, J. et al. The iPSYCH2015 Case-Cohort sample: updated directions for unravelling genetic and environmental architectures of severe mental disorders. Preprint at medRxiv https://doi.org/10.1101/2020.11.30.20237768 (2020).

  26. Pedersen, C. B. et al. The iPSYCH2012 case-cohort sample: new directions for unravelling genetic and environmental architectures of severe mental disorders. Mol. Psychiatry 23, 6–14 (2018).

  27. Mors, O., Perto, G. P. & Mortensen, P. B. The Danish psychiatric central research register. Scand. J. Public Health 39, 54–57 (2011).

    PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  29. 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 

  30. Yang, J., Lee, S. H., Goddard, M. E. & Visscher, P. M. Genome-wide complex trait analysis (GCTA): methods, data analyses, and interpretations. Methods Mol. Biol. 1019, 215–236 (2013).

    CAS  PubMed  Google Scholar 

  31. Stankiewicz, P. et al. Haploinsufficiency of the chromatin remodeler BPTF causes syndromic developmental and speech delay, postnatal microcephaly, and dysmorphic features. Am. J. Hum. Genet 101, 503–515 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Ansar, M. et al. Biallelic variants in LINGO1 are associated with autosomal recessive intellectual disability, microcephaly, speech and motor delay. Genet. Med. 20, 778–784 (2018).

    CAS  PubMed  Google Scholar 

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

    PubMed  Google Scholar 

  34. Duman, R. S., Shinohara, R., Fogaca, M. V. & Hare, B. Neurobiology of rapid-acting antidepressants: convergent effects on GluA1-synaptic function. Mol. Psychiatry 24, 1816–1832 (2019).

    PubMed  PubMed Central  Google Scholar 

  35. Hara, H. et al. TAK-653, an AMPA receptor potentiator with minimal agonistic activity, produces an antidepressant-like effect with a favorable safety profile in rats. Pharmacol. Biochem. Behav. 211, 173289 (2021).

    CAS  PubMed  Google Scholar 

  36. 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 

  37. Verduijn, J. et al. Using clinical characteristics to identify which patients with major depressive disorder have a higher genetic load for three psychiatric disorders. Biol. Psychiatry 81, 316–324 (2017).

    PubMed  Google Scholar 

  38. Zheng, J. et al. LD Hub: a centralized database and web interface to perform LD score regression that maximizes the potential of summary level GWAS data for SNP heritability and genetic correlation analysis. Bioinformatics 33, 272–279 (2017).

    CAS  PubMed  Google Scholar 

  39. Johnson, E. C. et al. A large-scale genome-wide association study meta-analysis of cannabis use disorder. Lancet Psychiatry 7, 1032–1045 (2020).

    PubMed  PubMed Central  Google Scholar 

  40. Zhou, H. et al. Genome-wide meta-analysis of problematic alcohol use in 435,563 individuals yields insights into biology and relationships with other traits. Nat. Neurosci. 23, 809–818 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Deak, J. D. & Johnson, E. C. Genetics of substance use disorders: a review. Psychol. Med. 51, 2181–2200 (2021).

  42. Gelernter, J. & Polimanti, R. Genetics of substance use disorders in the era of big data. Nat. Rev. Genet. 22, 712–729 (2021).

  43. Olvera, R. L. et al. Common genetic influences on depression, alcohol, and substance use disorders in Mexican-American families. Am. J. Med. Genet. B Neuropsychiatr. Genet. 156B, 561–568 (2011).

    CAS  PubMed  Google Scholar 

  44. Adams, M. J. et al. Genetic stratification of depression by neuroticism: revisiting a diagnostic tradition. Psychol. Med. 50, 2526–2535 (2020).

    PubMed  Google Scholar 

  45. Frei, O. et al. Bivariate causal mixture model quantifies polygenic overlap between complex traits beyond genetic correlation. Nat. Commun. 10, 2417 (2019).

    PubMed  PubMed Central  Google Scholar 

  46. Demontis, D. et al. Genome-wide analyses of ADHD identify 27 risk loci, refine the genetic architecture and implicate several cognitive domains. Nat. Genet. 55, 198–208 (2023).

  47. Werme, J., van der Sluis, S., Posthuma, D. & de Leeuw, C. A. An integrated framework for local genetic correlation analysis. Nat. Genet. 54, 274–282 (2022).

    CAS  PubMed  Google Scholar 

  48. Watanabe, K., Taskesen, E., van Bochoven, A. & Posthuma, D. Functional mapping and annotation of genetic associations with FUMA. Nat. Commun. 8, 1826 (2017).

    PubMed  PubMed Central  Google Scholar 

  49. Zhang, W. et al. Integrative transcriptome imputation reveals tissue-specific and shared biological mechanisms mediating susceptibility to complex traits. Nat. Commun. 10, 3834 (2019).

    PubMed  PubMed Central  Google Scholar 

  50. Wang, D. et al. Comprehensive functional genomic resource and integrative model for the human brain. Science 362, eaat8464 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Gandal, M. J. et al. Transcriptome-wide isoform-level dysregulation in ASD, schizophrenia, and bipolar disorder. Science 362, eaat8127 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Berisa, T. & Pickrell, J. K. Approximately independent linkage disequilibrium blocks in human populations. Bioinformatics 32, 283–285 (2016).

    CAS  PubMed  Google Scholar 

  53. Zeng, B. et al. Multi-ancestry eQTL meta-analysis of human brain identifies candidate causal variants for brain-related traits. Nat. Genet. 54, 161–169 (2022).

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

    PubMed  PubMed Central  Google Scholar 

  55. Dobbyn, A. et al. Landscape of conditional eQTL in dorsolateral prefrontal cortex and co-localization with schizophrenia GWAS. Am. J. Hum. Genet 102, 1169–1184 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Hormozdiari, F. et al. Colocalization of GWAS and eQTL signals detects target genes. Am. J. Hum. Genet 99, 1245–1260 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Meuleman, W. et al. Index and biological spectrum of human DNase I hypersensitive sites. Nature 584, 244–251 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Corces, M. R. et al. Single-cell epigenomic analyses implicate candidate causal variants at inherited risk loci for Alzheimer’s and Parkinson’s diseases. Nat. Genet. 52, 1158–1168 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. 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 

  60. Holmes, S. E. et al. Lower synaptic density is associated with depression severity and network alterations. Nat. Commun. 10, 1529 (2019).

    PubMed  PubMed Central  Google Scholar 

  61. Wang, Y. T. et al. Novel rapid-acting glutamatergic modulators: targeting the synaptic plasticity in depression. Pharmacol. Res. 171, 105761 (2021).

    CAS  PubMed  Google Scholar 

  62. Szebeni, A. et al. Shortened telomere length in white matter oligodendrocytes in major depression: potential role of oxidative stress. Int. J. Neuropsychopharmacol. 17, 1579–1589 (2014).

  63. Zhou, B., Zhu, Z., Ransom, B. R. & Tong, X. Oligodendrocyte lineage cells and depression. Mol. Psychiatry 26, 103–117 (2021).

    PubMed  Google Scholar 

  64. Leng, L. et al. Menin deficiency leads to depressive-like behaviors in mice by modulating astrocyte-mediated neuroinflammation. Neuron 100, 551–563 (2018).

    CAS  PubMed  Google Scholar 

  65. Satterthwaite, T. D. et al. Neuroimaging of the Philadelphia Neurodevelopmental Cohort. Neuroimage 86, 544–553 (2014).

    PubMed  Google Scholar 

  66. Calkins, M. E. et al. The Philadelphia Neurodevelopmental Cohort: constructing a deep phenotyping collaborative. J. Child Psychol. Psychiatry 56, 1356–1369 (2015).

    PubMed  PubMed Central  Google Scholar 

  67. Gur, R. C. et al. Age group and sex differences in performance on a computerized neurocognitive battery in children age 8-21. Neuropsychology 26, 251–265 (2012).

    PubMed  PubMed Central  Google Scholar 

  68. Wilkinson, G. S. & Robertson, G. J. Wide Range Achievement Test (WRAT4) (Pearson).

  69. Cullen, B. et al. Cognitive function and lifetime features of depression and bipolar disorder in a large population sample: cross-sectional study of 143,828 UK Biobank participants. Eur. Psychiatry 30, 950–958 (2015).

    CAS  PubMed  Google Scholar 

  70. Lim, J. et al. Sensitivity of cognitive tests in four cognitive domains in discriminating MDD patients from healthy controls: a meta-analysis. Int. Psychogeriatr. 25, 1543–1557 (2013).

    PubMed  Google Scholar 

  71. Snyder, H. R. Major depressive disorder is associated with broad impairments on neuropsychological measures of executive function: a meta-analysis and review. Psychol. Bull. 139, 81–132 (2013).

    PubMed  Google Scholar 

  72. Musliner, K. L. et al. Polygenic liability, stressful life events and risk for secondary-treated depression in early life: a nationwide register-based case-cohort study. Psychol. Med. 53, 217–226 (2023).

  73. Musliner, K. L. et al. Polygenic liability and recurrence of depression in patients with first-onset depression treated in hospital-based settings. JAMA Psychiatry 78, 792–795 (2021).

  74. Steffen, A., Nubel, J., Jacobi, F., Batzing, J. & Holstiege, J. Mental and somatic comorbidity of depression: a comprehensive cross-sectional analysis of 202 diagnosis groups using German nationwide ambulatory claims data. BMC Psychiatry 20, 142 (2020).

    PubMed  PubMed Central  Google Scholar 

  75. Richards, D. Prevalence and clinical course of depression: a review. Clin. Psychol. Rev. 31, 1117–1125 (2011).

    PubMed  Google Scholar 

  76. Green, E. K. et al. Variation at the GABAA receptor gene, Rho 1 (GABRR1) associated with susceptibility to bipolar schizoaffective disorder. Am. J. Med. Genet. B Neuropsychiatr. Genet. 153B, 1347–1349 (2010).

    PubMed  Google Scholar 

  77. Gulsuner, S. et al. Genetics of schizophrenia in the South African Xhosa. Science 367, 569–573 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Duman, R. S., Sanacora, G. & Krystal, J. H. Altered connectivity in depression: GABA and glutamate neurotransmitter deficits and reversal by novel treatments. Neuron 102, 75–90 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Selten, M., van Bokhoven, H. & Nadif Kasri, N. Inhibitory control of the excitatory/inhibitory balance in psychiatric disorders. F1000Res. 7, 23 (2018).

  81. Gao, R. & Penzes, P. Common mechanisms of excitatory and inhibitory imbalance in schizophrenia and autism spectrum disorders. Curr. Mol. Med. 15, 146–167 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Bruining, H. et al. Measurement of excitation-inhibition ratio in autism spectrum disorder using critical brain dynamics. Sci. Rep. 10, 9195 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Nelson, S. B. & Valakh, V. Excitatory/inhibitory balance and circuit homeostasis in autism spectrum disorders. Neuron 87, 684–698 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Sequeira, A. et al. Global brain gene expression analysis links glutamatergic and GABAergic alterations to suicide and major depression. PLoS ONE 4, e6585 (2009).

    PubMed  PubMed Central  Google Scholar 

  85. Merali, Z. et al. Dysregulation in the suicide brain: mRNA expression of corticotropin-releasing hormone receptors and GABA(A) receptor subunits in frontal cortical brain region. J. Neurosci. 24, 1478–1485 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Vinkers, C. H., Mirza, N. R., Olivier, B. & Kahn, R. S. The inhibitory GABA system as a therapeutic target for cognitive symptoms in schizophrenia: investigational agents in the pipeline. Expert Opin. Investig. Drugs 19, 1217–1233 (2010).

    CAS  PubMed  Google Scholar 

  87. Engin, E., Liu, J. & Rudolph, U. α2-containing GABA(A) receptors: a target for the development of novel treatment strategies for CNS disorders. Pharmacol. Ther. 136, 142–152 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Braat, S. & Kooy, R. F. The GABAA receptor as a therapeutic target for neurodevelopmental disorders. Neuron 86, 1119–1130 (2015).

    CAS  PubMed  Google Scholar 

  89. Benasi, G. et al. Benzodiazepines as a monotherapy in depressive disorders: a systematic review. Psychother. Psychosom. 87, 65–74 (2018).

    PubMed  Google Scholar 

  90. Fasipe, O. J., Agede, O. A. & Enikuomehin, A. C. Announcing the novel class of GABA-A receptor selective positive allosteric modulator antidepressants. Future Sci. OA 7, FSO654 (2020).

    PubMed  PubMed Central  Google Scholar 

  91. Hindley, G. et al. The shared genetic basis of mood instability and psychiatric disorders: a cross-trait genome-wide association analysis. Am. J. Med. Genet. B Neuropsychiatr. Genet. 189, 207–218 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Hujoel, M. L. A., Loh, P.-R., Neale, B. M. & Price, A. L. Incorporating family history of disease improves polygenic risk scores in diverse populations. Cell Genom. 2, 100152 (2022).

  93. Torkamani, A., Wineinger, N. E. & Topol, E. J. The personal and clinical utility of polygenic risk scores. Nat. Rev. Genet. 19, 581–590 (2018).

    CAS  PubMed  Google Scholar 

  94. Vieta, E. et al. Early intervention in bipolar disorder. Am. J. Psychiatry 175, 411–426 (2018).

    PubMed  Google Scholar 

  95. Lieberman, J. A., Small, S. A. & Girgis, R. R. Early detection and preventive intervention in schizophrenia: from fantasy to reality. Am. J. Psychiatry 176, 794–810 (2019).

    PubMed  Google Scholar 

  96. Carney, T. & Myers, B. Effectiveness of early interventions for substance-using adolescents: findings from a systematic review and meta-analysis. Subst. Abus. Treat. Prev. Policy 7, 25 (2012).

    Google Scholar 

  97. Kessler, R. C. & Price, R. H. Primary prevention of secondary disorders: a proposal and agenda. Am. J. Community Psychol. 21, 607–633 (1993).

    CAS  PubMed  Google Scholar 

  98. Polygenic Risk Score Task Force of the International Common Disease. Responsible use of polygenic risk scores in the clinic: potential benefits, risks and gaps. Nat. Med. 27, 1876–1884 (2021).

  99. Hollegaard, M. V. et al. Robustness of genome-wide scanning using archived dried blood spot samples as a DNA source. BMC Genet. 12, 58 (2011).

    PubMed  PubMed Central  Google Scholar 

  100. Borglum, A. D. et al. Genome-wide study of association and interaction with maternal cytomegalovirus infection suggests new schizophrenia loci. Mol. Psychiatry 19, 325–333 (2014).

  101. Illumina GenCall Data Analysis Software (Illumina, 2005).

  102. Korn, J. M. et al. Integrated genotype calling and association analysis of SNPs, common copy number polymorphisms and rare CNVs. Nat. Genet. 40, 1253–1260 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Goldstein, J. I. et al. zCall: a rare variant caller for array-based genotyping: genetics and population analysis. Bioinformatics 28, 2543–2545 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Lam, M. et al. RICOPILI: Rapid Imputation for COnsortias PIpeLIne. Bioinformatics 36, 930–933 (2020).

    CAS  PubMed  Google Scholar 

  105. Price, A. L. et al. Principal components analysis corrects for stratification in genome-wide association studies. Nat. Genet. 38, 904–909 (2006).

    CAS  PubMed  Google Scholar 

  106. Schork, A. J. et al. A genome-wide association study of shared risk across psychiatric disorders implicates gene regulation during fetal neurodevelopment. Nat. Neurosci. 22, 353–361 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Loh, P. R. et al. Reference-based phasing using the Haplotype Reference Consortium panel. Nat. Genet. 48, 1443–1448 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Iglesias, A. I. et al. Haplotype reference consortium panel: practical implications of imputations with large reference panels. Hum. Mutat. 38, 1025–1032 (2017).

    CAS  PubMed  Google Scholar 

  110. Price, A. L. et al. Long-range LD can confound genome scans in admixed populations. Am. J. Hum. Genet. 83, 132–135 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Zhou, W. et al. Efficiently controlling for case-control imbalance and sample relatedness in large-scale genetic association studies. Nat. Genet. 50, 1335–1341 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Levey, D. F. et al. Reproducible genetic risk loci for anxiety: results from approximately 200,000 participants in the Million Veteran Program. Am. J. Psychiatry 177, 223–232 (2020).

    PubMed  PubMed Central  Google Scholar 

  113. 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 

  114. Pasman, J. A. et al. GWAS of lifetime cannabis use reveals new risk loci, genetic overlap with psychiatric traits, and a causal influence of schizophrenia. Nat. Neurosci. 21, 1161–1170 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Walters, R. K. et al. Transancestral GWAS of alcohol dependence reveals common genetic underpinnings with psychiatric disorders. Nat. Neurosci. 21, 1656–1669 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Karlsson Linner, R. et al. Genome-wide association analyses of risk tolerance and risky behaviors in over 1 million individuals identify hundreds of loci and shared genetic influences. Nat. Genet. 51, 245–257 (2019).

    CAS  PubMed  Google Scholar 

  117. Liu, M. et al. Association studies of up to 1.2 million individuals yield new insights into the genetic etiology of tobacco and alcohol use. Nat. Genet. 51, 237–244 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Mullins, N. et al. Genome-wide association study of more than 40,000 bipolar disorder cases provides new insights into the underlying biology. Nat. Genet. 53, 817–829 (2021).

  119. Okbay, A. et al. Polygenic prediction of educational attainment within and between families from genome-wide association analyses in 3 million individuals. Nat. Genet. 54, 437–449 (2022).

  120. Nagel, M., Watanabe, K., Stringer, S., Posthuma, D. & van der Sluis, S. Item-level analyses reveal genetic heterogeneity in neuroticism. Nat. Commun. 9, 905 (2018).

    PubMed  PubMed Central  Google Scholar 

  121. Trubetskoy, V. et al. Mapping genomic loci implicates genes and synaptic biology in schizophrenia. Nature 604, 502–508 (2022).

  122. Bellenguez, C. et al. New insights into the genetic etiology of Alzheimer’s disease and related dementias. Nat. Genet. 54, 412–436 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. International League Against Epilepsy Consortium on Complex. Genome-wide mega-analysis identifies 16 loci and highlights diverse biological mechanisms in the common epilepsies. Nat. Commun. 9, 5269 (2018).

  124. Yengo, L. et al. A saturated map of common genetic variants associated with human height. Nature 610, 704–712 (2022).

  125. Schaid, D. J., Chen, W. & Larson, N. B. From genome-wide associations to candidate causal variants by statistical fine-mapping. Nat. Rev. Genet. 19, 491–504 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Kichaev, G. et al. Improved methods for multi-trait fine mapping of pleiotropic risk loci. Bioinformatics 33, 248–255 (2017).

    CAS  PubMed  Google Scholar 

  127. Kichaev, G. et al. Integrating functional data to prioritize causal variants in statistical fine-mapping studies. PLoS Genet. 10, e1004722 (2014).

    PubMed  PubMed Central  Google Scholar 

  128. Kichaev, G. & Pasaniuc, B. Leveraging functional-annotation data in trans-ethnic fine-mapping studies. Am. J. Hum. Genet. 97, 260–271 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Benner, C. et al. FINEMAP: efficient variable selection using summary data from genome-wide association studies. Bioinformatics 32, 1493–1501 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Bennett, D. A. et al. Religious orders study and rush memory and aging project. J. Alzheimer’s Dis. 64, S161–S189 (2018).

    Google Scholar 

  131. Consortium, G. T. et al. Genetic effects on gene expression across human tissues. Nature 550, 204–213 (2017).

    Google Scholar 

  132. Prive, F., Arbel, J. & Vilhjalmsson, B. J. LDpred2: better, faster, stronger. Bioinformatics 36, 5424–5431 (2020).

  133. Therneau, T. M. & Grambsch, P. M. Modeling Survival Data: Extending the Cox Model (Springer, 2000).

  134. A Package for Survival Analysis in R. R package v.3.2-13 (2021).

  135. Link, C. L. Confidence-intervals for the survival function using Cox proportional-hazard model with covariates. Biometrics 40, 601–610 (1984).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Klei, L., Kent, B. P., Melhem, N., Devlin, B. & Roeder, K. Gemtools: a fast and efficient approach to estimating genetic ancestry. Preprint at https://arxiv.org/abs/1104.1162 (2011).

  138. Ge, T., Chen, C. Y., Ni, Y., Feng, Y. A. & Smoller, J. W. Polygenic prediction via Bayesian regression and continuous shrinkage priors. Nat. Commun. 10, 1776 (2019).

    PubMed  PubMed Central  Google Scholar 

  139. Chang, C. C. et al. Second-generation PLINK: rising to the challenge of larger and richer datasets. GigaScience 4, 7 (2015).

    PubMed  PubMed Central  Google Scholar 

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

Download references

Acknowledgements

The iPSYCH team was supported by grants from the Lundbeck Foundation (R102-A9118, R155-2014-1724 and R248-2017-2003), the EU H2020 Program (grant no. 667302, ‘CoCA’), National Institutes of Health/National Institute of Mental Health (1U01MH109514-01 and 1R01MH124851-01 to A.D.B.) and the universities and university hospitals of Aarhus and Copenhagen. The Danish National Biobank resource was supported by the Novo Nordisk Foundation. High-performance computer capacity for the handling and statistical analysis of iPSYCH data on the GenomeDK HPC facility (https://genome.au.dk/) was provided by the Center for Genomics and Personalized Medicine and the Centre for Integrative Sequencing, iSEQ, Aarhus University, Denmark (grant to A.D.B.). We thank the research participants and employees of 23andMe, Inc. for making this work possible. We thank all members of the iPSYCH-Broad Consortium for their efforts and collaborative spirit. The team at the Center for Disease Neurogenomics at the Icahn School of Medicine at Mount Sinai was supported by the National Institutes of Health (K08MH122911 to G.V.; T32MH087004 to K.T.; and R01MH125246, R01AG067025, U01MH116442 and R01MH109677 to P.R.). D.F.L. was funded by a Veterans Affairs Office of Research and Development Career Development Award (IK2BX005058). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

T.D.A., M.I.K., J.G., G.V., K.T., E.T., T.T.N., J.N., K.V., D.F.L., J.B. and B.Z. were responsible for analysis. Sample and/or data provider and processing was carried out by J.B.G., D.D., A.R., G.A., M.B.H., P.Q., G.B.W., T.T., H.S., K.L.M., V.M.R., L.F., J.T., B.J.V., J.J.M., M.M., S.M., iPSYCH-Broad Consortium, E.A., K.S., M.M., T.W., D.M.H., P.B.M., M.B.S., J.G., I.H., P.R., M.J.D., O.M., A.P. and A.D.B. Supervision was carried out by P.Q., K.L.M., B.J.V., J.J.M, I.H., P.R., M.J.D., O.M., A.P. and A.D.B. Writing was the responsibility of T.D.A. and A.D.B. Study design and direction was the responsibility of A.D.B. All authors contributed to critical revision of the paper.

Corresponding authors

Correspondence to Thomas D. Als or Anders D. Børglum.

Ethics declarations

Competing interests

B.J.V. is a member of the advisory board for Allelica. D.D. has received a speaker fee from Takeda. B.W., T.T., H.S. and K.S. are employed at deCODE/Amgen. M.J.D. is a founder of Maze Therapeutics and is on the Scientific Advisory Board of RBNC Therapeutics. The other authors declare no competing interests.

Peer review

Peer review information

Nature Medicine thanks Gabriëlla Blokland, Gerome Breen and Steven Kushner for their contribution to the peer review of this work. Primary Handling Editor: Ming Yang, in collaboration with the Nature Medicine team.

Additional information

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

Extended data

Extended Data Fig. 1 Highlighted Regional Miami plots of GWAS and TWAS results.

corresponding to the genomic region of (a) GABRA1, (b) CYP7B1, (c) DCC, (d) CTTNBP2, (e) FURIN and (f) GIGYF2 genes/transcripts (1Mbp window from start site). Top panels: GWAS results (dots colored according to European ancestry linkage disequilibrium r2 with top SNP): The x-axis shows genomic position, and the y axis shows significance as –log10(P) of z statistics (two-sided nominal P values); blue line corresponds to P = 1 × 10−5, orange line to P = 5 × 10−8 (genome-wide significance). Bottom panels: TWAS results: The x-axis shows genomic position. The y axis shows significance as –log10(P) of z statistics (two-sided nominal P values) for genes represented by both gene expression and isoform expression. Black triangles facing upwards or downwards for a positive or negative association z-score (Wald test; two-sided P values) respectively (up- or down-regulation); transcripts with Bonferroni-adjusted (for all reliably imputed transcripts) P value < 0.1 are labeled; orange line corresponds to Bonferroni-adjusted P = 0.05. Each Bonferroni-significant transcript is connected with lines to the SNPs contributing to its transcriptomic imputation model; lines are gray when the SNPs have a P>1 × 10−5, blue when P < 1 × 10−5 but orange when P < 5 × 10−8. The SNPs that are above the blue line and contribute to the transcriptomic imputation models of significant transcripts are labeled. See Supplementary Fig. 10-1 to 10-88 and Supplementary Table 10.

Extended Data Fig. 2 MvPRS analyses of depression cases with/without anxiety (ANX).

Depression-subphenotype is shown on the x-axis (NDEPwoANX = 22114, NDEPwANX = 7044 and Nctrls = 38142). The slope (β) of the linear regression (95% CI) for each depression subphenotype is shown on the y axis. Significant difference between β for depression without/with an additional diagnosis is indicated with horizontal line with nominal two-sided P value above, that is the Wald test of equal group effect (see Supplementary Table 12a). Overall two-sided P value = 1.2 × 1021. Cases with BP were excluded. The polygenic risk scores analyzed are (a) PRS for depression (DEP-PRS), (b) PRS for anxiety (ANX-PRS), (c) PRS for bipolar disorder (BP-PRS), (d) PRS for schizophrenia (SZ-PRS), (e) PRS for ADHD (ADHD-PRS), (f) PRS for autism (ASD-PRS), (g) PRS for neuroticism (Neuroticism PRS), (h) PRS for substance use (SU-PRS), and (i) PRS for substance use disorder (SUD-PRS). See Supplementary Table 18b, 18c, Supplementary Fig. 19-1 and 19-2 for sex-stratified analyses.

Extended Data Fig. 3 MvPRS analyses of depression cases with/without bipolar disorder (BP).

Depression-subphenotype is shown on the x-axis (NDEPwoBP = 29158, DEPwBP, N = 1460 and Nctrls = 38200). The slope (β) of the linear regression (95% CI) for each depression subphenotype is shown on the y axis. Significant difference between β for depression without/with an additional diagnosis is indicated with horizontal line with nominal two-sided P value above, that is the Wald test of equal group effect (See Supplementary Table 12d). Overall two-sided P value = 1.5 × 1015. The polygenic risk scores analyzed are (a) PRS for depression (DEP-PRS), (b) PRS for anxiety (ANX-PRS), (c) PRS for bipolar disorder (BP-PRS), (d) PRS for schizophrenia (SZ-PRS), (e) PRS for ADHD (ADHD-PRS), (f) PRS for autism (ASD-PRS), (g) PRS for neuroticism (Neuroticism PRS), (h) PRS for substance use (SU-PRS), and (i) PRS for substance use disorder (SUD-PRS). See Supplementary Table 18e, S18f, Supplementary Fig. 19-1 and 19-2 for sex-stratified analyses.

Extended Data Fig. 4 MvPRS analyses of depression cases with/without schizophrenia (SZ).

Depression-subphenotype is shown on the x-axis (NDEPwoSZ = 25253, NDEPwSZ = 3905 and Nctrls = 38142). The slope (β) of the linear regression (95% CI) for each depression subphenotype is shown on the y axis. Significant difference between β for depression without/with an additional diagnosis is indicated with horizontal line with nominal two-sided P value above, that is the Wald test of equal group effect (see Supplementary Table 12g). Overall two-sided P value = 1.7 × 1015. Cases with BP were excluded. The polygenic risk scores analyzed are (a) PRS for depression (DEP-PRS), (b) PRS for anxiety (ANX-PRS), (c) PRS for bipolar disorder (BP-PRS), (d) PRS for schizophrenia (SZ-PRS), (e) PRS for ADHD (ADHD-PRS), (f) PRS for autism (ASD-PRS), (g) PRS for neuroticism (Neuroticism PRS), (h) PRS for substance use (SU-PRS), and (i) PRS for substance use disorder (SUD-PRS). See Supplementary Table 18h, 18i, Supplementary Fig. 19-1 and 19-2 for sex-stratified analyses.

Extended Data Fig. 5 MvPRS analyses of depression cases with/without substance use disorder (SUD).

Depression-subphenotype is shown on the x-axis (NDEPwoSUD = 25620, NDEPwSUD = 3538 and Nctrls = 38142). The slope (β) of the linear regression (95% CI) for each depression subphenotype is shown on the y axis. Significant difference between β for depression without/with an additional diagnosis is indicated with horizontal line with nominal two-sided P value above, that is the Wald test of equal group effect (see Supplementary Table 12j). Overall two-sided P value = 6.4 × 1097. Cases with BP were excluded. The polygenic risk scores analyzed are (a) PRS for depression (DEP-PRS), (b) PRS for anxiety (ANX-PRS), (c) PRS for bipolar disorder (BP-PRS), (d) PRS for schizophrenia (SZ-PRS), (e) PRS for ADHD (ADHD-PRS), (f) PRS for autism (ASD-PRS), (g) PRS for neuroticism (Neuroticism PRS), (h) PRS for substance use (SU-PRS), and (i) PRS for substance use disorder (SUD-PRS). See Supplementary Table 18k, 18l, Supplementary Fig. 19-1 and 19-2 for sex-stratified analyses.

Extended Data Fig. 6 Absolute risk and HRR of developing anxiety.

Left subpanels: Absolute risk (95%-CI) of developing anxiety since first depression episode for three groups of PRS deciles (1st, 2nd-to-9th and 10th) among 25124 depression cases with/without (NDEPwANX = 3010, NDEPwoANX = 22114) anxiety. The absolute risk (95%-CI) of anxiety in light blue for the iPSYCH2015 subcohort (random population sample) excluding all depression cases, aligned to match the endpoint for the depression-cohort. Right subpanels: HRRs (95%) for 1st and 10th decile using 2nd-to-9th decile as reference (see Supplementary Table 13a) in colors matching the absolute risks curves. For deciles of (a) Depression DEP-PRS, (b) Anxiety ANX-PRS, (c) Bipolar disorder BP-PRS, (d) Schizophrenia SZ-PRS, (e) ADHD-PRS, (f) Autism ASD-PRS, (g) Neuroticism PRS, (h) Substance Use SU-PRS, (i) Substance Use Disorder SUD-PRS and (j) sum of PRSs. The SUM-PRS was calculated by adding PRSs for multiple phenotypes weighted by log(OR) with the aim of optimizing prediction (see Methods for details). See Supplementary Fig. 20-120-2 and Supplementary Table 19b19c for sex-stratified analyses.

Extended Data Fig. 7 Absolute risk and HRR of transitioning into bipolar disorder.

Left subpanels: Absolute risk (95%-CI) of transitioning into bipolar disorder since first depression episode for three groups of PRS deciles (1st, 2nd-to-9th and 10th) among 30300 depression cases with/without (NDEPwBP = 1142, NDEPwoBP = 29158) anxiety. The absolute risk (95%-CI) of anxiety in light blue for the iPSYCH2015 subcohort (random population sample) excluding all depression cases, aligned to match the endpoint for the depression-cohort. Right subpanels: HRRs (95%) for 1st and 10th decile using 2nd-to-9th decile as reference (see Supplementary Table 14a) in colors matching the absolute risks curves. For deciles of (a) Depression DEP-PRS, (b) Anxiety ANX-PRS, (c) Bipolar disorder BP-PRS, (d) Schizophrenia SZ-PRS, (e) ADHD-PRS, (f) Autism ASD-PRS, (g) Neuroticism PRS, (h) Substance Use SU-PRS, (i) Substance Use Disorder SUD-PRS and (j) sum of PRSs. The SUM-PRS was calculated by adding PRSs for multiple phenotypes weighted by log(OR) with the aim of optimizing prediction (see Methods for details). See Supplementary Fig. 21-121-2 Supplementary Tables 20b20c for sex-stratified analyses.

Extended Data Fig. 8 Absolute risk and HRR of developing schizophrenia.

Left subpanels: Absolute risk (95%-CI) of developing schizophrenia since first depression episode for three groups of PRS deciles (1st, 2nd-to-9th and 10th) among 28714 depression cases with/without (NDEPwSZ = 1606, NDEPwoSZ = 27108) anxiety. The absolute risk (95%-CI) of anxiety in light blue for the iPSYCH2015 subcohort (random population sample) excluding all depression cases, aligned to match the endpoint for the depression-cohort. Right subpanels: HRRs (95%) for 1st and 10th decile using 2nd-to-9th decile as reference (see Supplementary Table 15a) in colors matching the absolute risks curves. For deciles of (a) Depression DEP-PRS, (b) Anxiety ANX-PRS, (c) Bipolar disorder BP-PRS, (d) Schizophrenia SZ-PRS, (e) ADHD-PRS, (f) Autism ASD-PRS, (g) Neuroticism PRS, (h) Substance Use SU-PRS, (i) Substance Use Disorder SUD-PRS and (j) sum of PRSs. The SUM-PRS was calculated by adding PRSs for multiple phenotypes weighted by log(OR) with the aim of optimizing prediction (see Methods for details). See Supplementary Fig. 22-122-2 and Supplementary Table 21b21c for sex-stratified analyses.

Extended Data Fig. 9 Absolute risk and HRR of developing SUD.

Left subpanels: Absolute risk (95%-CI) of developing SUD since first depression episode for three groups of PRS deciles (1st, 2nd-to-9th and 10th) among 27249 depression cases with/without (NDEPwSUD = 1629, NDEPwoSUD = 25620) anxiety. The absolute risk (95%-CI) of anxiety in light blue for the iPSYCH2015 subcohort (random population sample) excluding all depression cases, aligned to match the endpoint for the depression-cohort. Right subpanels: HRRs (95%) for 1st and 10th decile using 2nd-to-9th decile as reference (see Supplementary Table 16a) in colors matching the absolute risks curves. For deciles of (a) Depression DEP-PRS, (b) Anxiety ANX-PRS, (c) Bipolar disorder BP-PRS, (d) Schizophrenia SZ-PRS, (e) ADHD-PRS, (f) Autism ASD-PRS, (g) Neuroticism PRS, (h) Substance Use SU-PRS, (i) Substance Use Disorder SUD-PRS and (j) sum of PRSs. The SUM-PRS was calculated by adding PRSs for multiple phenotypes weighted by log(OR) with the aim of optimizing prediction (see Methods for details). See Supplementary Fig. 23-123-2 and Supplementary Tables 22b22c for sex-stratified analyses.

Extended Data Table 1 Number of comorbid diagnoses among depression subtypes: single-episode and recurrent depression

Supplementary information

Supplementary Information

Supplementary Tables 1–28 and Supplementary Figs. 16–23.

Reporting Summary

Supplementary Table 1

a, Independent index SNPs of the primary DEP meta-analysis. b, Results from the stepwise selection procedure selecting independently associated SNPs implemented in GCTA-COJO.

Supplementary Table 5

a, Genetic correlation (rG) of the primary and narrow DEP meta-analysis and DEP-narrow meta-analysis verus published non-UKB GWAS summary statistics available at LD Hub or locally. b, Genetic correlation (rG) of the primary and narrow DEP meta-analysis and DEP-narrow meta-analysis versus published UKB GWAS summary statistics available at LD Hub.

Supplementary Table 7

a, Datasets used in FUMA SNP2GENE analyses. b, Genome-wide-significant variants mapped to genes using FUMA. c, All candidate SNPs with annotations. d, Results from MAGMA gene-based association analysis.

Supplementary Table 8

a, List of parameters in the FUMA GENE2FUNC analysis. b, Results of the gene-to-function analysis in FUMA.

Supplementary Table 9

a, TWAS summary. b, List of Bonferroni-significant genes and transcripts by LD block. c, List of FDR significant genes. d, List of FDR significant transcripts.

Supplementary Table 13

Independent index SNPs of the narrow DEP GWAS meta-analysis.

Supplementary Table 15

Genetic correlations of other traits with DEP subtypes within iPSYCH2015. a, Based on non-UKB GWAS summary statistics available at LD Hub or locally. b, Based on UKB GWAS summary statistics available at LD Hub.

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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Als, T.D., Kurki, M.I., Grove, J. et al. Depression pathophysiology, risk prediction of recurrence and comorbid psychiatric disorders using genome-wide analyses. Nat Med 29, 1832–1844 (2023). https://doi.org/10.1038/s41591-023-02352-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-023-02352-1

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