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:

A genome-wide association study with 1,126,563 individuals identifies new risk loci for Alzheimer’s disease

An Author Correction to this article was published on 20 June 2022

An Author Correction to this article was published on 12 November 2021

This article has been updated

Abstract

Late-onset Alzheimer’s disease is a prevalent age-related polygenic disease that accounts for 50–70% of dementia cases. Currently, only a fraction of the genetic variants underlying Alzheimer’s disease have been identified. Here we show that increased sample sizes allowed identification of seven previously unidentified genetic loci contributing to Alzheimer’s disease. This study highlights microglia, immune cells and protein catabolism as relevant to late-onset Alzheimer’s disease, while identifying and prioritizing previously unidentified genes of potential interest. We anticipate that these results can be included in larger meta-analyses of Alzheimer’s disease to identify further genetic variants that contribute to Alzheimer’s pathology.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: A Manhattan plot of the meta-analysis results highlighting 38 loci, including seven previously unidentified regions.

Similar content being viewed by others

Data availability

Access to raw data can be requested via the Psychiatric Genomics Data Access portal (https://www.med.unc.edu/pgc/shared-methods/open-source-philosophy/), UK Biobank (www.ukbiobank.ac.uk), or 23andMe. Restriction of raw data is to protect the privacy of participants. Summary statistics from IGAP (https://web.pasteur-lille.fr/en/recherche/u744/igap/igap_download.php) and Finngen (https://www.finngen.fi/en/access_results) can be obtained from their respective online portals. Summary statistics from the meta-analysis excluding 23andMe are available at https://ctg.cncr.nl/software/summary_statistics. Access to the full set, including 23andMe results, can be obtained after approval from 23andMe is presented to the corresponding author. Approval can be obtained by completion of a Data Transfer Agreement. The Data Transfer Agreement exists to protect the privacy of 23andMe participants. Please visit https://research.23andme.com/dataset-access/ to initiate a request. Summary statistics of the primary microglia eQTLs are also available from EGA (accession no.: EGAD00001005736). MSigDB gene sets are available online (https://www.gsea-msigdb.org/gsea/msigdb/) and integrated in FUMA (https://fuma.ctglab.nl/).

Code availability

The code used to perform the analyses is available at https://github.com/dwightman/PGC-ALZ2. All software used in the analyses is freely available online.

Change history

References

  1. Bacigalupo, I. et al. A systematic review and meta-analysis on the prevalence of dementia in Europe: estimates from the highest-quality studies adopting the DSM IV diagnostic criteria. J. Alzheimers Dis. 66, 1471–1481 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Winblad, B. et al. Defeating Alzheimer’s disease and other dementias: a priority for European science and society. Lancet Neurol. 15, 455–532 (2016).

    Article  PubMed  Google Scholar 

  3. DeTure, M. A. & Dickson, D. W. The neuropathological diagnosis of Alzheimer’s disease. Mol. Neurodegener. 14, 32 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Gatz, M. et al. Heritability for Alzheimer’s disease: the study of dementia in Swedish twins. J. Gerontol. A Biol. Sci. Med. Sci. 52, M117–M125 (1997).

    Article  CAS  PubMed  Google Scholar 

  5. Gatz, M. et al. Role of genes and environments for explaining Alzheimer disease. Arch. Gen. Psychiatry 63, 168–174 (2006).

    Article  PubMed  Google Scholar 

  6. Zhang, Q. et al. Risk prediction of late-onset Alzheimer’s disease implies an oligogenic architecture. Nat. Commun. 11, 4799 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Holland, D. et al. The genetic architecture of human complex phenotypes is modulated by linkage disequilibrium and heterozygosity. Genetics 217, iyaa046 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Jansen, I. E. et al. Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer’s disease risk. Nat. Genet. 51, 404–413 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. 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 

  10. Lambert, J.-C. et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat. Genet. 45, 1452–1458 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. de Bakker, P. I. W. et al. Practical aspects of imputation-driven meta-analysis of genome-wide association studies. Hum. Mol. Genet. 17, R122–R128 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. 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 

  13. 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 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Kunkle, B. W. et al. Genetic meta-analysis of diagnosed Alzheimer’s disease identifies new risk loci and implicates Aβ, tau, immunity and lipid processing. Nat. Genet. 51, 414–430 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. 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  CAS  Google Scholar 

  16. Watanabe, K., Umićević Mirkov, M., de Leeuw, C. A., van den Heuvel, M. P. & Posthuma, D. Genetic mapping of cell type specificity for complex traits. Nat. Commun. 10, 3222 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Wishart, D. S. et al. DrugBank 5.0: a major update to the DrugBank database for 2018. Nucleic Acids Res. 46, D1074–D1082 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Shamilov, R. & Aneskievich, B. J. TNIP1 in autoimmune diseases: regulation of Toll-like receptor signaling. J. Immunol. Res. 2018, 3491269 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Cho, C. E. et al. A modular analysis of microglia gene expression, insights into the aged phenotype. BMC Genomics 20, 164 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Nho, K. et al. Association analysis of rare variants near the APOE region with CSF and neuroimaging biomarkers of Alzheimer’s disease. BMC Med. Genomics 10, 29 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Li, X. et al. Systematic analysis and biomarker study for Alzheimer’s disease. Sci. Rep. 8, 17394 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Marques-Coelho, D. et al. Differential transcript usage unravels gene expression alterations in Alzheimer’s disease human brains. NPJ Aging Mech. Dis. 7, 2 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Olah, M. et al. A transcriptomic atlas of aged human microglia. Nat. Commun. 9, 539 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Hickman, S. E. et al. The microglial sensome revealed by direct RNA sequencing. Nat. Neurosci. 16, 1896–1905 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Nam, K. N. et al. Effect of high fat diet on phenotype, brain transcriptome and lipidome in Alzheimer’s model mice. Sci. Rep. 7, 4307 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Oláh, J. et al. Interactions of pathological hallmark proteins: tubulin polymerization promoting protein/p25, beta-amyloid, and alpha-synuclein. J. Biol. Chem. 286, 34088–34100 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Mazaheri, F. et al. Distinct roles for BAI1 and TIM-4 in the engulfment of dying neurons by microglia. Nat. Commun. 5, 4046 (2014).

    Article  CAS  PubMed  Google Scholar 

  29. Ciani, M., Benussi, L., Bonvicini, C. & Ghidoni, R. Genome wide association study and next generation sequencing: a glimmer of light toward new possible horizons in frontotemporal dementia research. Front. Neurosci. 13, 506 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Li, Z. et al. The TMEM106B FTLD-protective variant, rs1990621, is also associated with increased neuronal proportion. Acta Neuropathol. 139, 45–61 (2020).

    Article  CAS  PubMed  Google Scholar 

  31. Prodan, C. I. et al. Coated-platelet levels and progression from mild cognitive impairment to Alzheimer disease. Neurology 76, 247–252 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Greaves, C. V. & Rohrer, J. D. An update on genetic frontotemporal dementia. J. Neurol. 266, 2075–2086 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Zhang, J. et al. Leukocyte immunoglobulin-like receptors in human diseases: an overview of their distribution, function, and potential application for immunotherapies. J. Leukoc. Biol. 102, 351–360 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Cao, Q. et al. Inhibiting amyloid-β cytotoxicity through its interaction with the cell surface receptor LilrB2 by structure-based design. Nat. Chem. 10, 1213–1221 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Liu, J. Z., Erlich, Y. & Pickrell, J. K. Case-control association mapping by proxy using family history of disease. Nat. Genet. 49, 325–331 (2017).

    Article  CAS  PubMed  Google Scholar 

  36. Schwabe, T., Srinivasan, K. & Rhinn, H. Shifting paradigms: the central role of microglia in Alzheimer’s disease. Neurobiol. Dis. 143, 104962 (2020).

    Article  CAS  PubMed  Google Scholar 

  37. Yao, D. W., O’Connor, L. J., Price, A. L. & Gusev, A. Quantifying genetic effects on disease mediated by assayed gene expression levels. Nat. Genet. 52, 626–633 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Winkler, T. W. et al. Quality control and conduct of genome-wide association meta-analyses. Nat. Protoc. 9, 1192–1212 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Kuhn, R. M., Haussler, D. & Kent, W. J. The UCSC genome browser and associated tools. Brief. Bioinform. 14, 144–161 (2013).

    Article  CAS  PubMed  Google Scholar 

  41. Ionita-Laza, I., Lee, S., Makarov, V., Buxbaum, J. D. & Lin, X. Sequence kernel association tests for the combined effect of rare and common variants. Am. J. Hum. Genet. 92, 841–853 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. 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  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  44. Sudlow, C. et al. UK Biobank: an open access resource for identifying the causes of a wide range of complex diseases of middle and old age. PLoS Med. 12, e1001779 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Liu, P.-P., Xie, Y., Meng, X.-Y. & Kang, J.-S. History and progress of hypotheses and clinical trials for Alzheimer’s disease. Signal Transduct. Target. Ther. 4, 29 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Marioni, R. E. et al. GWAS on family history of Alzheimer’s disease. Transl. Psychiatry 8, 99 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Desikan, R. S. et al. Genetic assessment of age-associated Alzheimer disease risk: development and validation of a polygenic hazard score. PLoS Med. 14, e1002258 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Jun, G. et al. A novel Alzheimer disease locus located near the gene encoding tau protein. Mol. Psychiatry 21, 108–117 (2016).

    Article  CAS  PubMed  Google Scholar 

  49. de Rojas, I. et al. Common variants in Alzheimer’s disease and risk stratification by polygenic risk scores. Nat. Commun. 12, 3417 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Schwartzentruber, J. et al. Genome-wide meta-analysis, fine-mapping and integrative prioritization implicate new Alzheimer’s disease risk genes. Nat. Genet. 53, 392–402 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. 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 

  52. 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 

  53. de Leeuw, C. A., Stringer, S., Dekkers, I. A., Heskes, T. & Posthuma, D. Conditional and interaction gene-set analysis reveals novel functional pathways for blood pressure. Nat. Commun. 9, 3768 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Hodge, R. D. et al. Conserved cell types with divergent features in human versus mouse cortex. Nature 573, 61–68 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Habib, N. et al. Massively parallel single-nucleus RNA-seq with DroNc-seq. Nat. Methods 14, 955–958 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Zhong, S. et al. A single-cell RNA-seq survey of the developmental landscape of the human prefrontal cortex. Nature 555, 524–528 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Enge, M. et al. Single-cell analysis of human pancreas reveals transcriptional signatures of aging and somatic mutation patterns. Cell 171, 321–330.e14 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Hochgerner, H. et al. STRT-seq-2i: dual-index 5′ single cell and nucleus RNA-seq on an addressable microwell array. Sci. Rep. 7, 16327 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Han, X. et al. Mapping the mouse cell atlas by Microwell-seq. Cell 172, 1091–1107.e17 (2018).

    Article  CAS  PubMed  Google Scholar 

  61. Zheng, G. X. Y. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. 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 

  63. Alasoo, K. et al. Shared genetic effects on chromatin and gene expression indicate a role for enhancer priming in immune response. Nat. Genet. 50, 424–431 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Chen, L. et al. Genetic drivers of epigenetic and transcriptional variation in human immune cells. Cell 167, 1398–1414.e24 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Jaffe, A. E. et al. Developmental and genetic regulation of the human cortex transcriptome illuminate schizophrenia pathogenesis. Nat. Neurosci. 21, 1117–1125 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Momozawa, Y. et al. IBD risk loci are enriched in multigenic regulatory modules encompassing putative causative genes. Nat. Commun. 9, 2427 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Fairfax, B. P. et al. Genetics of gene expression in primary immune cells identifies cell type-specific master regulators and roles of HLA alleles. Nat. Genet. 44, 502–510 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Fairfax, B. P. et al. Innate immune activity conditions the effect of regulatory variants upon monocyte gene expression. Science 343, 1246949 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Gutierrez-Arcelus, M. et al. Passive and active DNA methylation and the interplay with genetic variation in gene regulation. Elife 2, e00523 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Kasela, S. et al. Pathogenic implications for autoimmune mechanisms derived by comparative eQTL analysis of CD4+ versus CD8+ T cells. PLoS Genet. 13, e1006643 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Lepik, K. et al. C-reactive protein upregulates the whole blood expression of CD59—an integrative analysis. PLoS Comput. Biol. 13, e1005766 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Naranbhai, V. et al. Genomic modulators of gene expression in human neutrophils. Nat. Commun. 6, 7545 (2015).

    Article  PubMed  Google Scholar 

  73. Nédélec, Y. et al. Genetic ancestry and natural selection drive population differences in immune responses to pathogens. Cell 167, 657–669.e21 (2016).

    Article  PubMed  CAS  Google Scholar 

  74. Quach, H. et al. Genetic adaptation and Neandertal admixture shaped the immune system of human populations. Cell 167, 643–656.e17 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Schwartzentruber, J. et al. Molecular and functional variation in iPSC-derived sensory neurons. Nat. Genet. 50, 54–61 (2018).

    Article  CAS  PubMed  Google Scholar 

  76. Buil, A. et al. Gene-gene and gene-environment interactions detected by transcriptome sequence analysis in twins. Nat. Genet. 47, 88–91 (2015).

    Article  CAS  PubMed  Google Scholar 

  77. Võsa, U. et al. Unraveling the polygenic architecture of complex traits using blood eQTL metaanalysis. Preprint at bioRxiv https://doi.org/10.1101/447367 (2018).

  78. Westra, H.-J. et al. Systematic identification of trans eQTLs as putative drivers of known disease associations. Nat. Genet. 45, 1238–1243 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Zhernakova, D. V. et al. Identification of context-dependent expression quantitative trait loci in whole blood. Nat. Genet. 49, 139–145 (2017).

    Article  CAS  PubMed  Google Scholar 

  80. Ng, B. et al. An xQTL map integrates the genetic architecture of the human brain’s transcriptome and epigenome. Nat. Neurosci. 20, 1418–1426 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. 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 

  83. Wallace, C. Eliciting priors and relaxing the single causal variant assumption in colocalisation analyses. PLoS Genet. 16, e1008720 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Kerimov, N. et al. eQTL Catalogue: a compendium of uniformly processed human gene expression and splicing QTLs. Preprint at bioRxiv https://doi.org/10.1101/2020.01.29.924266 (2020).

  85. Young, A. M. H. et al. A map of transcriptional heterogeneity and regulatory variation in human microglia. Nat. Genet. 53, 861–868 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Wang, G., Sarkar, A., Carbonetto, P. & Stephens, M. A simple new approach to variable selection in regression, with application to genetic fine mapping. J. R. Stat. Soc. B Stat. Methodol. 82, 1273–1300 (2020).

    Article  Google Scholar 

  87. Benner, C. et al. Prospects of fine-mapping trait-associated genomic regions by using summary statistics from genome-wide association studies. Am. J. Hum. Genet. 101, 539–551 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. R Core Team. R: A Language and Environment for Statistical Computing (R Foundation for Statistical Computing, 2017).

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Lawrence, M. et al. Software for computing and annotating genomic ranges. PLoS Comput. Biol. 9, e1003118 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. 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  CAS  Google Scholar 

  92. O’Leary, N. A. et al. Reference sequence (RefSeq) database at NCBI: current status, taxonomic expansion, and functional annotation. Nucleic Acids Res. 44, D733–D745 (2016).

    Article  PubMed  CAS  Google Scholar 

  93. Hadley, W. ggplot2: Elegant Graphics for Data Analysis (Springer-Verlag, New York).

Download references

Acknowledgements

We thank all the participants included in this study, including the participants from Finngen, GR@CE, IGAP, UKB, DemGene, TwinGene, STSA, the Gothenburg H70 Birth Cohort Studies and Clinical AD from Sweden, ANMerge, BioVU, 23andMe, HUNT, and deCODE. We thank the research participants from 23andMe who made this study possible. We thank the participants of the Norwegian Dementia Genetics Network (DemGene). This work was supported by BRAINSCAPES: A Roadmap from Neurogenetics to Neurobiology (grant no. 024.004.012), and a European Research Council advanced grant (grant no. ERC-2018-AdG GWAS2FUNC 834057 (to D.P.)). This work was supported by the Research Council of Norway (RCN: 248980, 248778, 223273), Norwegian Regional Health Authorities, Norwegian Health Association (22731, EU JPND: PMI-AD RCN 311993); and National Institutes of Health, National Institute on Aging R01 AG08724, R01 AG17561, R01 AG028555 and R01 AG060470. We thank the International Genomics of Alzheimer’s Project (IGAP) for providing summary results data for these analyses. The investigators within IGAP contributed to the design and implementation of IGAP and/or provided data but did not participate in analysis or writing of this report. IGAP was made possible by the generous participation of the control subjects, the patients and their families. The iSelect chips were funded by the French National Foundation on Alzheimer’s disease and related disorders. EADI was supported by the LABEX (laboratory of excellence program investment for the future) DISTALZ grant, Inserm, Institut Pasteur de Lille, Université de Lille 2 and the Lille University Hospital. GERAD/PERADES was supported by the Medical Research Council (grant no. 503480), Alzheimer’s Research UK (grant no. 503176), the Wellcome Trust (grant no. 082604/2/07/Z) and German Federal Ministry of Education and Research (BMBF): Competence Network Dementia (CND) (grant nos. 01GI0102, 01GI0711, 01GI0420). CHARGE was partly supported by the NIH/NIA grant R01 AG033193 and the NIA AG081220 and AGES contract N01–AG–12100, the NHLBI grant R01 HL105756, the Icelandic Heart Association and the Erasmus Medical Center and Erasmus University. ADGC was supported by the NIH/NIA grants U01 AG032984, U24 AG021886, U01 AG016976 and the Alzheimer’s Association grant ADGC–10–196728.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

D.P. and O.A.A. conceived of the study. D.P.W. performed the meta-analysis and follow-up analyses. I.E.J., J.E.S., D.P. and O.A.A. supervised analyses. I.E.J. and J.E.S. generated the UKB data. S. Bahrami and A.A.S. helped with the study design. A.H.S., C.W., J.B.N., L.G.F., M.E.G., K.H., T.W.M. and M.B.J. contributed to the organization of the HUNT data. B.S.W., A.E.M., O.K.D., G.B., I.B., E.S., S. Børte, L.F.T., W.Z., J.-A.Z., S.B.S., G.S. and L.M.P. contributed to the methods and analysis of the HUNT data. D.A., E.S., O.A.A., A.R. and G.S. collected and analyzed the DemGene data. S.K., K.B., A.Z., I. Skoog, M.W. and H.Z. financed and collected the Gothenburg H70 Birth Cohort Studies and Clinical AD Sweden data, and A.Z. processed and coordinated the analysis. A.H. contributed to the IGAP and ANMerge data. P.P. provided ANMerge data. D.H. provided power estimates. R.D., L.V., the 23andMe Research Team, J.M.S., L.K.D., N.L.P., C.A.R., I.K.K., S.M., H.S., S.T., P.V.J., J.S., L.A., P.S., I. Saltvedt, I.U., S.D., T.F., S.R. and K.S. analyzed and provided data. D.P.W. wrote the first draft of the manuscript. All authors critically reviewed the paper

Corresponding author

Correspondence to Danielle Posthuma.

Ethics declarations

Competing interests

H.Z. has served at scientific advisory boards for Denali, Roche Diagnostics, Wave, Samumed, Siemens Healthineers, Pinteon Therapeutics and CogRx; has given lectures in symposia sponsored by Fujirebio, Alzecure and Biogen; and is a co-founder of Brain Biomarker Solutions in Gothenburg AB (BBS), which is a part of the GU Ventures Incubator Program (outside submitted work). K.B. has served as a consultant, at advisory boards, or at data monitoring committees for Abcam, Axon, Biogen, JOMDD/Shimadzu, Julius Clinical, Lilly, MagQu, Novartis, Roche Diagnostics and Siemens Healthineers and is a co-founder of Brain Biomarker Solutions in Gothenburg AB (BBS), which is a part of the GU Ventures Incubator Program. O.A.A. is a consultant to HealthLytix and received speaker’s honorarium from Lundbeck and Sunovion. J.B.N. is employed by Regeneron Pharmaceuticals, Inc. T.W.M. has received speaker’s honorarium from Roche. All other authors declare no competing interests.

Additional information

Peer review information Nature Genetics thanks the anonymous reviewers for their contribution to the peer review of this work. Peer reviewer reports are available.

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

Supplementary information

Supplementary Information

Supplementary Figs. 1–20, Results, Methods, References and further acknowledgements.

Reporting Summary

Peer Review File

Supplementary Tables

Supplementary Tables 1–17.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wightman, D.P., Jansen, I.E., Savage, J.E. et al. A genome-wide association study with 1,126,563 individuals identifies new risk loci for Alzheimer’s disease. Nat Genet 53, 1276–1282 (2021). https://doi.org/10.1038/s41588-021-00921-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-021-00921-z

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