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:

Genome-wide association analyses identify 95 risk loci and provide insights into the neurobiology of post-traumatic stress disorder

Abstract

Post-traumatic stress disorder (PTSD) genetics are characterized by lower discoverability than most other psychiatric disorders. The contribution to biological understanding from previous genetic studies has thus been limited. We performed a multi-ancestry meta-analysis of genome-wide association studies across 1,222,882 individuals of European ancestry (137,136 cases) and 58,051 admixed individuals with African and Native American ancestry (13,624 cases). We identified 95 genome-wide significant loci (80 new). Convergent multi-omic approaches identified 43 potential causal genes, broadly classified as neurotransmitter and ion channel synaptic modulators (for example, GRIA1, GRM8 and CACNA1E), developmental, axon guidance and transcription factors (for example, FOXP2, EFNA5 and DCC), synaptic structure and function genes (for example, PCLO, NCAM1 and PDE4B) and endocrine or immune regulators (for example, ESR1, TRAF3 and TANK). Additional top genes influence stress, immune, fear and threat-related processes, previously hypothesized to underlie PTSD neurobiology. These findings strengthen our understanding of neurobiological systems relevant to PTSD pathophysiology, while also opening new areas for investigation.

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: Data sources and analyses in PTSD Freeze 3.
Fig. 2: GWAS meta-analyses in European and multi-ancestry individuals identify a total of 95 PTSD risk loci.
Fig. 3: Manhattan plots of PTSD associations in multi-omic analyses.
Fig. 4: Gene prioritization in PTSD loci.
Fig. 5: PRS analysis for PTSD across different datasets and ancestries.

Similar content being viewed by others

Data availability

Summary statistics for PGC-PTSD Freeze 3 will be made available upon publication under the accession ID ptsd2024 via the PGC website (https://pgc.unc.edu/for-researchers/download-results/). Access to study-level summary statistics and genotype data can be applied by using the PGC data access portal (https://pgc.unc.edu/for-researchers/data-access-committee/data-access-portal/).

Code availability

Analysis code is made available at GitHub (https://github.com/nievergeltlab/freeze3_gwas) and Zenodo (https://doi.org/10.5281/zenodo.10182702)118.

References

  1. Koenen, K. C. et al. Posttraumatic stress disorder in the World Mental Health Surveys. Psychol. Med. 47, 2260–2274 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Davis, L. L. et al. The economic burden of posttraumatic stress disorder in the United States from a societal perspective. J. Clin. Psychiatry 83, 21m14116 (2022).

    Article  PubMed  Google Scholar 

  3. Ferland-Beckham, C. et al. Systematic review and methodological considerations for the use of single prolonged stress and fear extinction retention in rodents. Front. Behav. Neurosci. 15, 652636 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ressler, K. J. et al. Post-traumatic stress disorder: clinical and translational neuroscience from cells to circuits. Nat. Rev. Neurol. 18, 273–288 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. McClellan France, J. & Jovanovic, T. Human fear neurobiology reimagined: can brain-derived biotypes predict fear-based disorders after trauma? Neurosci. Biobehav. Rev. 144, 104988 (2023).

    Article  PubMed  Google Scholar 

  6. Dunsmoor, J. E., Cisler, J. M., Fonzo, G. A., Creech, S. K. & Nemeroff, C. B. Laboratory models of post-traumatic stress disorder: the elusive bridge to translation. Neuron 110, 1754–1776 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Bassil, K. et al. In vitro modeling of the neurobiological effects of glucocorticoids: a review. Neurobiol. Stress 23, 100530 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Seah, C. et al. Modeling gene × environment interactions in PTSD using human neurons reveals diagnosis-specific glucocorticoid-induced gene expression. Nat. Neurosci. 25, 1434–1445 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Kremen, W. S., Koenen, K. C., Afari, N. & Lyons, M. J. Twin studies of posttraumatic stress disorder: differentiating vulnerability factors from sequelae. Neuropharmacology 62, 647–653 (2012).

    Article  CAS  PubMed  Google Scholar 

  10. Wolf, E. J. et al. A classical twin study of PTSD symptoms and resilience: evidence for a single spectrum of vulnerability to traumatic stress. Depress. Anxiety 35, 132–139 (2018).

    Article  CAS  PubMed  Google Scholar 

  11. Nievergelt, C. M. et al. International meta-analysis of PTSD genome-wide association studies identifies sex- and ancestry-specific genetic risk loci. Nat. Commun. 10, 4558 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Maihofer, A. X. et al. Enhancing discovery of genetic variants for posttraumatic stress disorder through integration of quantitative phenotypes and trauma exposure information. Biol. Psychiatry 91, 626–636 (2022).

    Article  PubMed  Google Scholar 

  13. Stein, M. B. et al. Genome-wide association analyses of post-traumatic stress disorder and its symptom subdomains in the Million Veteran Program. Nat. Genet. 53, 174–184 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Wendt, F. R. et al. The relationship of attention-deficit/hyperactivity disorder with posttraumatic stress disorder: a two-sample Mendelian randomization and population-based sibling comparison study. Biol. Psychiatry 93, 362–369 (2023).

    Article  PubMed  Google Scholar 

  15. Polimanti, R. et al. Understanding the comorbidity between posttraumatic stress severity and coronary artery disease using genome-wide information and electronic health records. Mol. Psychiatry 27, 3961–3969 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Campbell-Sills, L. et al. Dissecting the heterogeneity of posttraumatic stress disorder: differences in polygenic risk, stress exposures, and course of PTSD subtypes. Psychol. Med. 52, 1–9 (2021).

    PubMed  Google Scholar 

  17. Choi, K. W. et al. Prospective study of polygenic risk, protective factors, and incident depression following combat deployment in US Army soldiers. Psychol. Med. 50, 737–745 (2020).

    Article  PubMed  Google Scholar 

  18. Lobo, J. J. et al. Polygenic risk scoring to assess genetic overlap and protective factors influencing posttraumatic stress, depression, and chronic pain after motor vehicle collision trauma. Transl. Psychiatry 11, 359 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Roberts, A. L., Gilman, S. E., Breslau, J., Breslau, N. & Koenen, K. C. Race/ethnic differences in exposure to traumatic events, development of post-traumatic stress disorder, and treatment-seeking for post-traumatic stress disorder in the United States. Psychol. Med. 41, 71–83 (2011).

    Article  CAS  PubMed  Google Scholar 

  20. Bassett, D., Buchwald, D. & Manson, S. Posttraumatic stress disorder and symptoms among American Indians and Alaska Natives: a review of the literature. Soc. Psychiatry Psychiatr. Epidemiol. 49, 417–433 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Logue, M. W. et al. The Psychiatric Genomics Consortium Posttraumatic Stress Disorder Workgroup: posttraumatic stress disorder enters the age of large-scale genomic collaboration. Neuropsychopharmacology 40, 2287–2297 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Holland, D. et al. Beyond SNP heritability: polygenicity and discoverability of phenotypes estimated with a univariate Gaussian mixture model. PLoS Genet. 16, e1008612 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  26. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. De Bakker, P. I. & Raychaudhuri, S. Interrogating the major histocompatibility complex with high-throughput genomics. Hum. Mol. Genet. 21, R29–R36 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Rentzsch, P., Witten, D., Cooper, G. M., Shendure, J. & Kircher, M. CADD: predicting the deleteriousness of variants throughout the human genome. Nucleic Acids Res. 47, D886–D894 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. Boyle, A. P. et al. Annotation of functional variation in personal genomes using RegulomeDB. Genome Res. 22, 1790–1797 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Weissbrod, O. et al. Functionally informed fine-mapping and polygenic localization of complex trait heritability. Nat. Genet. 52, 1355–1363 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Liberzon, A. et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 1, 417–425 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. La Manno, G. et al. Molecular diversity of midbrain development in mouse, human, and stem cells. Cell 167, 566–580 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Barbeira, A. N. et al. Exploiting the GTEx resources to decipher the mechanisms at GWAS loci. Genome Biol. 22, 49 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  37. Pathak, G. A. et al. Genetically regulated multi-omics study for symptom clusters of posttraumatic stress disorder highlights pleiotropy with hematologic and cardio-metabolic traits. Mol. Psychiatry 27, 1394–1404 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Waszczuk, M. A. et al. Discovery and replication of blood-based proteomic signature of PTSD in 9/11 responders. Transl. Psychiatry 13, 8 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Wingo, T. S. et al. Integrating human brain proteomes with genome-wide association data implicates novel proteins in post-traumatic stress disorder. Mol. Psychiatry 27, 3075–3084 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Sun, B. B. et al. Plasma proteomic associations with genetics and health in the UK Biobank. Nature 622, 329–338 (2022).

    Article  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Romero, C. et al. Exploring the genetic overlap between twelve psychiatric disorders. Nat. Genet. 54, 1795–1802 (2022).

    Article  CAS  PubMed  Google Scholar 

  43. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Watson, H. J. et al. Genome-wide association study identifies eight risk loci and implicates metabo-psychiatric origins for anorexia nervosa. Nat. Genet. 51, 1207–1214 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Otowa, T. et al. Meta-analysis of genome-wide association studies of anxiety disorders. Mol. Psychiatry 21, 1391–1399 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. International Obsessive Compulsive Disorder Foundation Genetics Collaborative (IOCDF-GC) & OCD Collaborative Genetics Association Studies (OCGAS) Revealing the complex genetic architecture of obsessive-compulsive disorder using meta-analysis. Mol. Psychiatry 23, 1181–1188 (2018).

  52. Yu, D. et al. Interrogating the genetic determinants of Tourette’s syndrome and other tic disorders through genome-wide association studies. Am. J. Psychiatry 176, 217–227 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  53. 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).

    Article  CAS  PubMed  Google Scholar 

  54. Bycroft, C. et al. The UK Biobank resource with deep phenotyping and genomic data. Nature 562, 203–209 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Zoellner, L. A., Roy-Byrne, P. P., Mavissakalian, M. & Feeny, N. C. Doubly randomized preference trial of prolonged exposure versus sertraline for treatment of PTSD. Am. J. Psychiatry 176, 287–296 (2019).

    Article  PubMed  Google Scholar 

  56. Bullman, T. A. & Kang, H. K. Posttraumatic stress disorder and the risk of traumatic deaths among Vietnam veterans. J. Nerv. Ment. Dis. 182, 604–610 (1994).

    Article  CAS  PubMed  Google Scholar 

  57. Clover, K., Carter, G. L. & Whyte, I. M. Posttraumatic stress disorder among deliberate self-poisoning patients. J. Trauma Stress 17, 509–517 (2004).

    Article  PubMed  Google Scholar 

  58. Gradus, J. L. et al. Posttraumatic stress disorder and gastrointestinal disorders in the Danish population. Epidemiology 28, 354–360 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Brady, K. T., Killeen, T. K., Brewerton, T. & Lucerini, S. Comorbidity of psychiatric disorders and posttraumatic stress disorder. J. Clin. Psychiatry 61, 22–32 (2000).

    PubMed  Google Scholar 

  60. Kind, S. & Otis, J. D. The interaction between chronic pain and PTSD. Curr. Pain Headache Rep. 23, 91 (2019).

    Article  PubMed  Google Scholar 

  61. Nishimi, K. et al. Post-traumatic stress disorder and risk for hospitalization and death following COVID-19 infection. Transl. Psychiatry 12, 482 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Roberts, A. L., Kubzansky, L. D., Chibnik, L. B., Rimm, E. B. & Koenen, K. C. Association of posttraumatic stress and depressive symptoms with mortality in women. JAMA Netw. Open 3, e2027935 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Schlenger, W. E. et al. A prospective study of mortality and trauma-related risk factors among a nationally representative sample of Vietnam veterans. Am. J. Epidemiol. 182, 980–990 (2015).

    PubMed  Google Scholar 

  64. Martin, A. R. et al. Human demographic history impacts genetic risk prediction across diverse populations. Am. J. Hum. Genet. 100, 635–649 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Panagiotou, O. A., Willer, C. J., Hirschhorn, J. N. & Ioannidis, J. P. The power of meta-analysis in genome-wide association studies. Annu. Rev. Genomics Hum. Genet. 14, 441–465 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Huggins, A. et al. Smaller total and subregional cerebellar volumes in posttraumatic stress disorder: a mega-analysis by the ENIGMA-PGC PTSD workgroup. Biol. Psychiatry 93, S44 (2023).

    Article  Google Scholar 

  67. Girgenti, M. J. et al. Transcriptomic organization of the human brain in post-traumatic stress disorder. Nat. Neurosci. 24, 24–33 (2021).

    Article  CAS  PubMed  Google Scholar 

  68. Logue, M. W. et al. Gene expression in the dorsolateral and ventromedial prefrontal cortices implicates immune-related gene networks in PTSD. Neurobiol. Stress 15, 100398 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Jaffe, A. E. et al. Decoding shared versus divergent transcriptomic signatures across cortico-amygdala circuitry in PTSD and depressive disorders. Am. J. Psychiatry 179, 673–686 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Chatzinakos, C. et al. Single-nucleus transcriptome profiling of dorsolateral prefrontal cortex: mechanistic roles for neuronal gene expression, including the 17q21.31 locus, in PTSD stress response. Am. J. Psychiatry 180, 739–754 (2023).

    Article  PubMed  Google Scholar 

  71. Kessler, R. C., Sonnega, A., Bromet, E., Hughes, M. & Nelson, C. B. Posttraumatic stress disorder in the national comorbidity survey. Arch. Gen. Psychiatry 52, 1048–1060 (1995).

    Article  CAS  PubMed  Google Scholar 

  72. Ravi, M., Stevens, J. S. & Michopoulos, V. Neuroendocrine pathways underlying risk and resilience to PTSD in women. Front. Neuroendocrinol. 55, 100790 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Hodes, G. E. & Epperson, C. N. Sex differences in vulnerability and resilience to stress across the life span. Biol. Psychiatry 86, 421–432 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Gelernter, J. et al. Genome-wide association study of post-traumatic stress disorder reexperiencing symptoms in >165,000 US veterans. Nat. Neurosci. 22, 1394–1401 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Nachtigall, E. G., de Freitas, J. D. R., de, C. M. J. & Furini, C. R. G. Role of hippocampal Wnt signaling pathways on contextual fear memory reconsolidation. Neuroscience 524, 108–119 (2023).

    Article  CAS  PubMed  Google Scholar 

  76. Lv, T. et al. Electroacupuncture alleviates PTSD-like behaviors by modulating hippocampal synaptic plasticity via Wnt/β-catenin signaling pathway. Brain Res. Bull. 202, 110734 (2023).

    Article  CAS  PubMed  Google Scholar 

  77. Herrero, M. J. et al. Sex-specific social behavior and amygdala proteomic deficits in Foxp2+/− mutant mice. Front. Behav. Neurosci. 15, 706079 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Dalvie, S. et al. Genomic influences on self-reported childhood maltreatment. Transl. Psychiatry 10, 38 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Grotzinger, A. D. et al. Genetic architecture of 11 major psychiatric disorders at biobehavioral, functional genomic and molecular genetic levels of analysis. Nat. Genet. 54, 548–559 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Kessler, R. C., Chiu, W. T., Demler, O., Merikangas, K. R. & Walters, E. E. Prevalence, severity, and comorbidity of 12-month DSM-IV disorders in the national comorbidity survey replication. Arch. Gen. Psychiatry 62, 617–627 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  81. Breen, G. et al. Translating genome-wide association findings into new therapeutics for psychiatry. Nat. Neurosci. 19, 1392–1396 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Stein, M. B. & Rothbaum, B. O. 175 years of progress in PTSD therapeutics: learning from the past. Am. J. Psychiatry 175, 508–516 (2018).

    Article  PubMed  Google Scholar 

  83. Mahoney, C. T., Moshier, S. J., Keane, T. M. & Marx, B. P. Heightened healthcare utilization & risk of mental disorders among Veterans with comorbid opioid use disorder & posttraumatic stress disorder. Addict. Behav. 112, 106572 (2021).

    Article  PubMed  Google Scholar 

  84. Upadhyay, J. et al. Neurocircuitry basis of the opioid use disorder-post-traumatic stress disorder comorbid state: conceptual analyses using a dimensional framework. Lancet Psychiatry 9, 84–96 (2022).

    Article  PubMed  Google Scholar 

  85. Lam, M. et al. RICOPILI: rapid imputation for COnsortias PIpeLIne. Bioinformatics 36, 930–933 (2020).

    Article  CAS  PubMed  Google Scholar 

  86. McCarthy, S. et al. A reference panel of 64,976 haplotypes for genotype imputation. Nat. Genet. 48, 1279–1283 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Chen, C. Y. et al. Improved ancestry inference using weights from external reference panels. Bioinformatics 29, 1399–1406 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  89. Zhou, X. & Stephens, M. Genome-wide efficient mixed-model analysis for association studies. Nat. Genet. 44, 821–824 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Chen, W. M., Manichaikul, A. & Rich, S. S. A generalized family-based association test for dichotomous traits. Am. J. Hum. Genet. 85, 364–376 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Loh, P.-R. et al. Efficient Bayesian mixed-model analysis increases association power in large cohorts. Nat. Genet. 47, 284–290 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Mbatchou, J. et al. Computationally efficient whole-genome regression for quantitative and binary traits. Nat. Genet. 53, 1097–1103 (2021).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Pruim, R. J. et al. LocusZoom: regional visualization of genome-wide association scan results. Bioinformatics 26, 2336–2337 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Auton, A. et al. A global reference for human genetic variation. Nature 526, 68–74 (2015).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  99. GTEx Consortium The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science 369, 1318–1330 (2020).

    Article  Google Scholar 

  100. 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 

  101. Hoffman, G. E. et al. CommonMind Consortium provides transcriptomic and epigenomic data for schizophrenia and bipolar disorder. Sci. Data 6, 180 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Paola, G.-R. et al. Using three-dimensional regulatory chromatin interactions from adult and fetal cortex to interpret genetic results for psychiatric disorders and cognitive traits. Preprint at bioRxiv https://doi.org/10.1101/406330 (2019).

  104. Schmitt, A. D. et al. A compendium of chromatin contact maps reveals spatially active regions in the human genome. Cell Rep. 17, 2042–2059 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Wang, K., Li, M. & Hakonarson, H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 38, e164 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  106. Zou, Y., Carbonetto, P., Wang, G. & Stephens, M. Fine-mapping from summary data with the “Sum of Single Effects” model. PLoS Genet. 18, e1010299 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Bryois, J. et al. Cell-type-specific cis-eQTLs in eight human brain cell types identify novel risk genes for psychiatric and neurological disorders. Nat. Neurosci. 25, 1104–1112 (2022).

    Article  CAS  PubMed  Google Scholar 

  108. Chatzinakos, C. et al. TWAS pathway method greatly enhances the number of leads for uncovering the molecular underpinnings of psychiatric disorders. Am. J. Med. Genet. B 183, 454–463 (2020).

    Article  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  110. Koopmans, F. et al. SynGO: an evidence-based, expert-curated knowledge base for the synapse. Neuron 103, 217–234 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Gaspar, H. A. & Breen, G. Drug enrichment and discovery from schizophrenia genome-wide association results: an analysis and visualisation approach. Sci. Rep. 7, 12460 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Freshour, S. L. et al. Integration of the Drug–Gene Interaction Database (DGIdb 4.0) with open crowdsource efforts. Nucleic Acids Res. 49, D1144–D1151 (2021).

    Article  CAS  PubMed  Google Scholar 

  113. Roth, B. L., Lopez, E., Patel, S. & Kroeze, W. K. The multiplicity of serotonin receptors: uselessly diverse molecules or an embarrassment of riches? Neuroscientist 6, 252–262 (2000).

    Article  CAS  Google Scholar 

  114. Mendez, D. et al. ChEMBL: towards direct deposition of bioassay data. Nucleic Acids Res. 47, D930–D940 (2019).

    Article  CAS  PubMed  Google Scholar 

  115. Sheils, T. K. et al. TCRD and Pharos 2021: mining the human proteome for disease biology. Nucleic Acids Res. 49, D1334–D1346 (2021).

    Article  CAS  PubMed  Google Scholar 

  116. Yoo, M. et al. DSigDB: drug signatures database for gene set analysis. Bioinformatics 31, 3069–3071 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Maihofer, A.X. nievergeltlab/PTSDF3: Release V0.99. Zenodo https://doi.org/10.5281/zenodo.10182702 (2023).

Download references

Acknowledgements

Major financial support for the PTSD-PGC was provided by the National Institute of Mental Health (NIMH; R01MH106595 (to K.C.K., C.M.N., K.J.R. and M.B.S.), R01MH124847 (to C.M.N.) and R01MH124851 (to A.D.B., L.K.D. and K.C.K.)), the Stanley Center for Psychiatric Research at the Broad Institute and Cohen Veterans Bioscience. Statistical analyses were carried out on the NL Genetic Cluster computer (URL) hosted by SURFsara. Genotyping of samples was supported in part through the Stanley Center for Psychiatric Genetics at the Broad Institute of MIT and Harvard. This research has been conducted using the UKB resource under application 41209. This work would not have been possible without the contributions of the investigators who comprise the PGC-PTSD working group, and especially the more than 1,307,247 research participants worldwide who shared their life experiences and biological samples with PGC-PTSD investigators. We thank A.E. Aiello, B. Bradley, A. Gautam, R. Hammamieh, M. Jett, M.J. Lyons, D. Maurer, M.R. Mavissakalian and the late C.R. Erbes and R.E. McGlinchey for their contributions to this study.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

E.G.A., S.-A.B., C.-Y.C., K.W.C., J.R.I.C., N.P.D., L.E.D., K.C.K., A.X.M., R.A.M., C.M.N., R.P., K.J.R. and M.B.S. were the members of the PGC-PTSD writing group. A.B.A., S.B. Andersen, P.A.A., A.E.A.-K., S.B. Austin, E.A., D.B., D.G.B., J.C.B., S. Belangero, C. Benjet, J.M.B., L.J.B., J.I.B., G.B., R.B., A.D.B., J.R.C., C.S.C., L.K.B., J.D., D.L.D., T.d.-C., K.D., G.D., A.D.-K., N.F., L.A.F., A.F., N.C.F., B.G., J.G., E.G., C.F.G., A.G.U., M.A.H., A.C.H., V.H., I.B.H., D.M.H., K. Hveem, M. Jakovljević, A.J., I.J., T.J., K.-I.K., M.L.K., R.C.K., N.A.K., K.C.K., R.K., H.R.K., W.S.K., B.R.L., K.L., I.L., B.L., C.M., N.G.M., K.A.M., S.A.M., S.E.M., D.M., W.P.M., M.W.M., C.P.M., O.M., P.B.M, E.C.N., C.M.N., M.N., S.B.N., N.R.N., P.M.P., A.L.P., R.H.P., M.A.P., B.P., A.P., K.J.R., V.R., P.R.B., K.R., H.R., G.S., S. Seedat, J.S. Seng, A.K.S., S.R.S., D.J.S., M.B.S., R.J.U., U.V., S.J.H.v.R., E.V., J.V., Z.W., M.W., H.W., T.W., M.A.W., D.E.W., C.W., R.M.Y., H.Z., L.A.Z. and J.-A.Z. were the principal investigators or co-principal investigators of contributing studies. A.B.A., P.A.A., A.E.A.-K., S.B. Austin, J.C.B., S. Belangero, C. Benjet, J.M.B., L.J.B., G.B., A.D.B., C.S.C., J.D., T.d.-C., A.F., N.C.F., J.D.F., C.E.F., E.G., C.F.G., M.H., M.A.H., A.C.H., V.H., I.B.H., D.M.H., K. Hveem, T.J., N.A.K., K.C.K., R.K., W.S.K., B.R.L., B.L., C.M., N.G.M., K.A.M., S.A.M., S.E.M., J.M., W.P.M., M.W.M., C.P.M., O.M., P.B.M, E.C.N., C.M.N., M.N., N.R.N., H.K.O., M.A.P., B.P., K.J.R., B.O.R., G.S., M.S., A.K.S., S.R.S., M.H.T., R.J.U., U.V., E.V., J.V., Z.W., M.W., T.W., M.A.W., D.E.W., R.Y., R.M.Y. and L.A.Z. obtained funding for studies. C.A., P.A.A., E.A., D.B., D.G.B., J.C.B., L.B., L.J.B., E.A.B., R.B., A.C.B., A.D.B., S. Børte, L.C., J.R.C., K.W.C., L.K.B., M.F.D., T.d.-C., S.G.D., G.D., A.D.-K., N.F., N.C.F., J.D.F., C.E.F., S.G., E.G., A.G.U., S.B.G., L.G., C.G., V.H., D.M.H., M. Jakovljević, A.J., G.D.J., M.L.K., A.K., N.A.K., N.K., R.K., W.S.K., B.R.L., L.A.M.L., K.L., C.E.L., B.L., J.L.M.-K., S.A.M., P.B.M., H.K.O., P.M.P., M.S.P., E.S.P., A.L.P., M.P., R.H.P., M.A.P., B.P., A.P., B.O.R., A.O.R., G.S., L.S., J.S. Seng, C.M.S., S. Stensland, M.H.T., W.K.T., E.T., M.U., U.V., L.L.v.d.H., E.V., Z.W., Y.W., T.W., D.E.W., B.S.W., S.W., E.J.W., R.Y., K.A.Y. and L.A.Z. were responsible for clinical aspects of studies. O.A.A., P.A.A., S.B. Austin, D.G.B., S. Belangero, L.J.B., R.B., R.A.B., A.D.B., J.R.C., J.M.C.-d.-A., S.Y.C., S.A.P.C., A.M.D., L.K.B., D.L.D., A.E., N.C.F., D.F., C.E.F., S.G., B.G., S.M.J.H., D.M.H., L.M.H., K. Hveem, A.J., I.J., M.L.K., J.L.K., R.C.K., A.P.K., R.K., W.S.K., L.A.M.L., K.L., D.F.L., C.E.L., I.L., B.L., M.K.L., S.M., G.A.M., K.M., A.M., K.A.M., S.E.M., J.M., L.M., O.M., P.B.M., M.N., S.B.N., N.R.N., M.O., P.M.P., M.S.P., E.S.P., A.L.P., M.P., R.H.P., M.A.P., K.J.R., V.R., P.R.B., A. Rung, G.S., L.S., S.E.S., M.S., C.S., S. Seedat, J.S. Seng, D. Silove, J.W.S., S.R.S., M.B.S., A.K.T., E.T., U.V., L.L.v.d.H., M.V.H., M.W., T.W., D.E.W., S.W., K.A.Y., C.C.Z., G.C.Z., L.A.Z. and J.-A.Z. contributed to data collection. A.E.A.-K., A. Batzler, C. Bergner, A. Brandolino, S. Børte, C.C., C.-Y.C., S.A.P.C., J.R.I.C., L.C.-C., B.J.C., S.D., S.G.D., A.D., L.E.D., C.F., M.E.G., B.G., S.B.G., S.D.G., C.G., S.H., E.M.H., K. Hogan, H.H., G.D.J., K.K., P.-F.K., D.F.L., M.W.L., A.L, Y.L., A.X.M., S.M., C.M., D.M., J.M., V.M., E.A.M., M.S.M., C.M.N., G.A.P., M.P., X.-J.Q., A.R., A.L.R., S.S.d.V., C.S., A.S., C.M.S., S. Stensland, J.S.S., J.A.S., F.R.W., B.S.W., Y. Xia, Y. Xiong and C.C.Z. conducted statistical analysis. A.E.A.-K., A. Batzler, M.P.B., S. Børte, C.C., C.-Y.C., J.R.I.C., N.P.D., C.D.P., S.G.D., A.D., H.E., M.E.G., K. Hogan, H.H., K.K., P.-F.K., D.F.L., S.D.L., A.L, A.X.M., G.A.M., D.M., J.M., V.M., E.A.M., G.A.P., A.R., A.S., J.S.S., F.R.W., B.S.W., C.W., Y. Xia, Y. Xiong and C.C.Z. conducted bioinformatics analysis. M.P.B., J.B.-G., M.B.-H., N.P.D., T.d.-C., F.D., A.D., K.D., H.E., L.G., M.A.H., J.J., P.-F.K., S.D.L., J.J.L., I.K., J.M., L.M., K.J.R., B.P.F.R., S.S.d.V., A.S., C.H.V. and D.E.W. conducted genomics studies. M.H. and M.Z were the members of the PGC-PTSD management group.

Corresponding author

Correspondence to Caroline M. Nievergelt.

Ethics declarations

Competing interests

L.J.B. is listed as an inventor on Issued US Patent 8,080,371, ‘Markers for Addiction’ covering the use of certain SNPs in determining the diagnosis, prognosis and treatment of addiction. C.-Y.C. and H.R. are employees of Biogen. A.M.D. holds equity in CorTechs Labs and serves on the Scientific Advisory Board of Human Longevity and the Mohn Medical Imaging and Visualization Center. A.M.D. receives funding through research grants with General Electric Healthcare. C.F. was a speaker for Janssen in 2021. I.B.H. is the codirector, Health and Policy at the Brain and Mind Center (BMC) University of Sydney; the BMC operates early intervention youth services at Camperdown under contract to headspace; and is the Chief Scientific Advisor to, and a 3.2% equity shareholder in, InnoWell Pty; InnoWell was formed by the University of Sydney (45% equity) and PwC (Australia; 45% equity) to deliver the AU$30 million Australian Government-funded Project Synergy. H.H. received consultancy fees from Ono Pharmaceutical and an honorarium from Xian Janssen Pharmaceutical. In the past 3 years, R.C.K. was a consultant for Cambridge Health Alliance, Canandaigua VA Medical Center, Holmusk, Partners Healthcare, RallyPoint Networks and Sage Therapeutics. He has stock options in Cerebral, Mirah, PYM, Roga Sciences and Verisense Health. L.A.M.L. reports spousal IP payments from Vanderbilt University for technology licensed to Acadia Pharmaceuticals unrelated to the present work. C.M. has served on advisory boards of Receptor Life Sciences, Otsuka Pharmaceuticals and Roche Products Limited and has received support from the National Institute on Alcohol Abuse and Alcoholism, NIMH, Department of Defense-CDMRP (US Army Research Office) DARPA, Bank of America Foundation, Brockman Foundation, Cohen Veterans Bioscience, Cohen Veterans Network, McCormick Foundation, Home Depot Foundation, New York City Council, New York State Health, Mother Cabrini Foundation, Tilray Pharmaceuticals and Ananda Scientific. P.M.P. received payment or honoraria for lectures and presentations in educational events for Sandoz, Daiichi Sankyo, Eurofarma, Abbot, Libbs, Instituto Israelita de Pesquisa e Ensino Albert Einstein, Instituto D’Or de Pesquisa e Ensino. R.P. has been paid for his editorial work on the journal Complex Psychiatry and received a research grant outside the scope of this study from Alkermes. J.W.S. is a member of the Scientific Advisory Board of Sensorium Therapeutics (with equity) and has received grant support from Biogen; and is the principal investigator of a collaborative study of the genetics of depression and BPD sponsored by 23andMe for which 23andMe provides analysis time as in-kind support but no payments. M.B.S. has in the past 3 years received consulting income from Acadia Pharmaceuticals, Aptinyx, atai Life Sciences, BigHealth, Biogen, Bionomics, BioXcel Therapeutics, Boehringer Ingelheim, Clexio, Eisai, EmpowerPharm, Engrail Therapeutics, Janssen, Jazz Pharmaceuticals, NeuroTrauma Sciences, PureTech Health, Sage Therapeutics, Sumitomo Pharma and Roche/Genentech; has stock options in Oxeia Biopharmaceuticals and EpiVario; has been paid for his editorial work on Depression and Anxiety (Editor-in-Chief), Biological Psychiatry (Deputy Editor) and UpToDate (Coeditor-in-Chief for Psychiatry); has also received research support from NIH, Department of Veterans Affairs and the Department of Defense; and is on the scientific advisory board for the Brain and Behavior Research Foundation and the Anxiety and Depression Association of America. In the past 3 years, D.J.S. has received consultancy honoraria from Discovery Vitality, Johnson & Johnson, Kanna, L’Oreal, Lundbeck, Orion, Sanofi, Servier, Takeda and Vistagen. M.L.K. reports unpaid membership on the Scientific Committee for the International Society for the Study of Trauma and Dissociation. The other authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks Jean-Baptiste Pingault and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

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 Comparison of the genetic architecture of PTSD in the three main data sources.

Quantification of polygenicity and polygenic overlap in the three main data subsets based on (1) symptom scores in clinical studies and cohorts assessed on a variety of instruments in Freeze 2.5 (yellow; 26,080 cases and 192,966 controls), (2) PCL (for DSM-IV) based symptom scores in the MVP (red; 32,372 cases and 154,317 controls) and (3) ICD9/10 codes in EHR studies (blue; 78,684 cases and 738,463 controls) indicate a similar genetic architecture. The circles on the top half of the plot depict univariate MiXeR estimates of the total polygenicity for each data subset. Numbers within circles indicate polygenicity values, expressed as the number of variants (in thousands, with s.e. in parenthesis) necessary to explain 90% of SNP-based heritability (h2SNP). h2SNP estimates are written in the boxes at the bottom of the circles. The Euler diagrams on the bottom half of the plot depict bivariate MiXeR estimates of the polygenic overlap between data subsets. Values in the overlapping part of the Euler diagrams denote shared polygenicity and values on the non-overlapping parts note dataset-specific polygenicity. Genetic correlations (rg) between dataset pairs are noted in the boxes below the Euler diagrams. Arrowed lines are drawn between univariate and bivariate results to indicate which dataset pairs are being evaluated.

Extended Data Fig. 2 Manhattan plot of the PTSD GWAS meta-analysis in individuals of European ancestry (EA).

Results of the EA GWAS meta-analysis (137,136 PTSD cases, 1,085,746 controls) identifying 81 genome-wide significant PTSD loci. The y-axis refers to the −log10 P value from two-sided z-tests for effect estimates for a meta-analysis using a sample size weighted fixed-effects model. Circle colors alternate between chromosomes: even chromosomes are colored blue and odd chromosomes are colored black. The horizontal red bar indicates genome-wide significant associations (P < 5 × 10−8).

Extended Data Fig. 3 Significant PTSD gene sets.

MAGMA gene-set analysis using the Molecular Signatures Database (MSigDB) identifies 12 significant gene sets. The dotted line indicates significance adjusted for the number of comparisons (P < 0.05/15,483 gene sets). Bars depict −log10 P values from one-sided t-tests for enrichment. Corresponding gene-set names are indicated to the left of bars. Terms are clustered and colored according to their Gene Ontology term category (biological processes, yellow; molecular function, blue; cellular component, red).

Extended Data Fig. 4 MAGMA tissue enrichment analysis.

MAGMA gene-property analysis in 53 specific tissue types from GTEx v8 shows enrichment of PTSD-related genes in 13 brain tissue types and in the pituitary. Bars depict −log10 P values from one-sided t-tests for enrichment. Corresponding tissue names are indicated below bars. The dotted horizontal line indicates statistical significance adjusted for the number of comparisons (P < 0.05/53). Significant tissues are colored red.

Extended Data Fig. 5 MAGMA cell-type enrichment analysis in midbrain.

MAGMA gene-property analysis of 25 midbrain cell types (GSE76381) indicates enrichment of GABAergic neurons, GABAergic neuroblasts and mediolateral neuroblasts. Vertical bars depict −log10 P values from one-sided t-tests for enrichment. Significant cell types are colored blue and gray if not. The dotted horizontal line indicates statistical significance adjusted for the number of comparisons (P < 0.05/25). The asterisk (*) indicates that GABAergic neurons remained significant in stepwise conditional analysis of the other significant cell types. Abbreviations: Gaba, GABAergic neurons; NbGaba, neuroblast gabaergic; NbML1-5, mediolateral neuroblasts; DA0-2, dopaminergic neurons; Sert, serotonergic neurons; RN, red nucleus; Rgl 1-3, radial glia-like cells; NbM, medial neuroblasts; OPC, oligodendrocyte precursor cells; ProgFPL, progenitor lateral floorplate; OMTN, oculomotor and trochlear nucleus; Endo, endothelial cells; ProgM, progenitor midline; NProg, neuronal progenitor; ProgBP, progenitor basal plate; Mgl, microglia; ProgFPM, progenitor medial floorplate; Peric, pericytes.

Extended Data Fig. 6 PTSD genes in SynGO.

Sunburst plots show enrichment of PTSD-related genes in SynGO cellular components. The synapse is at the center ring, pre- and post-synaptic locations are at the first rings, and child terms are in subsequent outer rings. a, Enrichment test results for all 415 genes mapped to PTSD GWAS loci by FUMA from one of three gene-mapping strategies (positional, expression quantitative trait loci and chromatin interaction mapping). b, Enrichment test results for 43 genes prioritized into tier 1 using a gene prioritization strategy. Plots are colored by −log10 Q-value (see color code in the bar at left) from enrichment of PTSD genes relative to a brain-expressed background set.

Extended Data Fig. 7 Genetic correlations and polygenic overlap between PTSD and other psychiatric disorders.

a, Genetic correlations (rg) with standard error between PTSD and 11 other psychiatric disorders are indicated by circles that are drawn along the x-axis. Red dots indicate SNP-based heritability (h2SNP) z-score >6 in the psychiatric disorder GWAS and colored gray to indicate z-score <6 (rg estimates may be unreliable). The first author and publication year of source summary data are noted in parenthesis following the disorder name. b, Quantification of the polygenic overlap between PTSD and other psychiatric disorders. Euler diagrams depict Bivariate MiXeR analysis of PTSD (blue circles) and bipolar disorder (BIP), major depression (MDD) and schizophrenia (SCZ) (red circles). Values in the overlapping part of the Euler diagrams denote shared polygenicity (expressed as the number of influential variants, in thousands, with s.e. in parenthesis), and values in the non-overlapping part indicate dataset-specific variation. rg between dataset pairs are noted in the boxes below the Euler plots. Abbreviations: ADHD, attention deficit hyperactive disorder; Alc. dep., alcohol dependence; BIP, bipolar disorder; MDD, major depression; OCD, obsessive compulsive disorder; SCZ, schizophrenia.

Supplementary information

Supplementary Information

Supplementary Note and Figs. 1–10.

Reporting Summary

Peer Review File

Supplementary Tables

Supplementary Tables 1–31.

Supplementary Data 1

Regional association plots for each significant locus from the EA GWAS meta-analysis.

Supplementary Data 2

Forest plots for each of the 81 index variants of the GWAS loci identified in the EA GWAS meta-analysis.

Supplementary Data 3

Circos plots of chromatin interactions and eQTLs for each chromosome with PTSD risk loci based on the EA GWAS meta-analysis.

Supplementary Data 4

Gene prioritization scores and rankings for all 415 protein-coding genes mapped to risk loci.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nievergelt, C.M., Maihofer, A.X., Atkinson, E.G. et al. Genome-wide association analyses identify 95 risk loci and provide insights into the neurobiology of post-traumatic stress disorder. Nat Genet (2024). https://doi.org/10.1038/s41588-024-01707-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41588-024-01707-9

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