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:

Genetics of the human microglia regulome refines Alzheimer’s disease risk loci

Abstract

Microglia are brain myeloid cells that play a critical role in neuroimmunity and the etiology of Alzheimer’s disease (AD), yet our understanding of how the genetic regulatory landscape controls microglial function and contributes to AD is limited. Here, we performed transcriptome and chromatin accessibility profiling in primary human microglia from 150 donors to identify genetically driven variation and cell-specific enhancer–promoter (E-P) interactions. Integrative fine-mapping analysis identified putative regulatory mechanisms for 21 AD risk loci, of which 18 were refined to a single gene, including 3 new candidate risk genes (KCNN4, FIBP and LRRC25). Transcription factor regulatory networks captured AD risk variation and identified SPI1 as a key putative regulator of microglia expression and AD risk. This comprehensive resource capturing variation in the human microglia regulome provides insights into the etiology of neurodegenerative disease.

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: Chromatin accessibility landscape in human microglia and AD predisposition.
Fig. 2: Transcriptional regulation by OCRs.
Fig. 3: Genetic regulation of chromatin accessibility in human microglia.
Fig. 4: Integration of AD etiologic landscape with genetic regulation of transcriptional and chromatin accessibility in microglia.
Fig. 5: TF binding landscape in microglia integrating AD genetics.

Similar content being viewed by others

Data availability

The genotypes, omics data and metadata are available via the AD Knowledge Portal (https://adknowledgeportal.org). The AD Knowledge Portal is a platform for accessing data, analyses and tools generated by the Accelerating Medicines Partnership (AMP-AD) Target Discovery Program and other National Institute on Aging-supported programs to enable open-science practices and accelerate translational learning. The data, analyses and tools are shared early in the research cycle without a publication embargo on secondary use. Data are available for general research use according to the requirements for data access and data attribution (https://adknowledgeportal.org/DataAccess/Instructions). For access to content described in this paper, see https://doi.org/10.7303/syn26207321, including the derived data available for open distribution: processed ATAC-seq and RNA-seq data, OCR information and annotation, QTL summary and fine-mapping results and colocalization results.

TF motifs used in MotifBreakR analyses use JASPAR2016 (http://jaspar.genereg.net). ATAC-seq data were filtered with DAC Blacklisted/Exclusion List Regions (doi: 10.17989/ENCSR636HFF). Lopes et al. microglia eQTL data are available from National Institute on Aging Genetics of Alzheimer’s Disease Data Storage Site (NIAGADS) at https://dss.niagads.org/datasets/ng00105/ under accession no. NG00105.v1 at https://dss.niagads.org/datasets/ng00105/). For Alasoo et al. caQTL and eQTL data, raw RNA-seq and ATAC-seq data are available from ENA (ERP020977) and EGA (EGAS00001002236), and processed read counts are available from Zenodo (doi: 10.5281/zenodo.259661). Young et al. microglia eQTL data are available under managed access from the EGA, upon approval by the Wellcome Sanger Institute Data Access Committee (https://ega-archive.org/datasets/EGAD00001005736).

Code availability

Code used throughout this study for analyses and visualization is available at https://doi.org/10.7303/syn26207321. Additional code may be available upon reasonable request from the corresponding authors.

References

  1. Salter, M. W. & Stevens, B. Microglia emerge as central players in brain disease. Nat. Med. 23, 1018–1027 (2017).

    Article  CAS  PubMed  Google Scholar 

  2. Bartels, T., De Schepper, S. & Hong, S. Microglia modulate neurodegeneration in Alzheimer’s and Parkinson’s diseases. Science 370, 66–69 (2020).

    Article  CAS  PubMed  Google Scholar 

  3. Song, W. M. & Colonna, M. The identity and function of microglia in neurodegeneration. Nat. Immunol. 19, 1048–1058 (2018).

    Article  CAS  PubMed  Google Scholar 

  4. Nott, A. et al. Brain cell type-specific enhancer-promoter interactome maps and disease-risk association. Science 366, 1134–1139 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Hauberg, M. E. et al. Common schizophrenia risk variants are enriched in open chromatin regions of human glutamatergic neurons. Nat. Commun. 11, 5581 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. 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 

  7. 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 

  8. Lopes, K. et al. Genetic analysis of the human microglial transcriptome across brain regions, aging and disease pathologies. Nat. Genet. 54, 4–17 (2022).

    Article  CAS  PubMed  Google Scholar 

  9. Gosselin, D. et al. An environment-dependent transcriptional network specifies human microglia identity. Science 356, eaal3222 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Novikova, G. et al. Integration of Alzheimer’s disease genetics and myeloid genomics identifies disease risk regulatory elements and genes. Nat. Commun. 12, 1610 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Morabito, S. et al. Single-nucleus chromatin accessibility and transcriptomic characterization of Alzheimer’s disease. Nat. Genet. 53, 1143–1155 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Liu, X. et al. Functional architectures of local and distal regulation of gene expression in multiple human tissues. Am. J. Hum. Genet. 100, 605–616 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. 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 

  14. Fulco, C. P. et al. Activity-by-contact model of enhancer-promoter regulation from thousands of CRISPR perturbations. Nat. Genet. 51, 1664–1669 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Bendl, J. et al. The three-dimensional landscape of chromatin accessibility in Alzheimer’s disease. Preprint at bioRxiv https://doi.org/10.1101/2021.01.11.426303 (2021).

  16. Nasser, J. et al. Genome-wide enhancer maps link risk variants to disease genes. Nature 593, 238–243 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. 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 

  19. Bryois, J. et al. Evaluation of chromatin accessibility in prefrontal cortex of individuals with schizophrenia. Nat. Commun. 9, 3121 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Han, B. & Eskin, E. Interpreting meta-analyses of genome-wide association studies. PLoS Genet. 8, e1002555 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Hormozdiari, F., Kostem, E., Kang, E. Y., Pasaniuc, B. & Eskin, E. Identifying causal variants at loci with multiple signals of association. Genetics 198, 497–508 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kim-Hellmuth, S. et al. Cell type-specific genetic regulation of gene expression across human tissues. Science 369, eaaz8528 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Zhou, J. & Troyanskaya, O. G. Predicting effects of noncoding variants with deep learning-based sequence model. Nat. Methods 12, 931–934 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Zhou, J. et al. Deep learning sequence-based ab initio prediction of variant effects on expression and disease risk. Nat. Genet. 50, 1171–1179 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Bentsen, M. et al. ATAC-seq footprinting unravels kinetics of transcription factor binding during zygotic genome activation. Nat. Commun. 11, 4267 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Huang, K.-L. et al. A common haplotype lowers PU.1 expression in myeloid cells and delays onset of Alzheimer’s disease. Nat. Neurosci. 20, 1052–1061 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. 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 

  28. Giambartolomei, C. et al. A Bayesian framework for multiple trait colocalization from summary association statistics. Bioinformatics 34, 2538–2545 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Xiao, Q. et al. Role of phosphatidylinositol clathrin assembly lymphoid-myeloid leukemia (PICALM) in intracellular amyloid precursor protein (APP) processing and amyloid plaque pathogenesis. J. Biol. Chem. 287, 21279–21289 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lachmann, A. et al. Geneshot: search engine for ranking genes from arbitrary text queries. Nucleic Acids Res. 47, W571–W577 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Yi, M. et al. KCa3.1 constitutes a pharmacological target for astrogliosis associated with Alzheimer’s disease. Mol. Cell. Neurosci. 76, 21–32 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Jin, L.-W. et al. Repurposing the KCa3.1 inhibitor senicapoc for Alzheimer’s disease. Ann. Clin. Transl. Neurol. 6, 723–738 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Rahman, M. R. et al. Network-based approach to identify molecular signatures and therapeutic agents in Alzheimer’s disease. Comput. Biol. Chem. 78, 431–439 (2019).

    Article  CAS  PubMed  Google Scholar 

  34. Acquaah-Mensah, G. K. & Taylor, R. C. Brain in situ hybridization maps as a source for reverse-engineering transcriptional regulatory networks: Alzheimer’s disease insights. Gene 586, 77–86 (2016).

    Article  CAS  PubMed  Google Scholar 

  35. Xu, J. et al. IL-4/STAT6 signaling facilitates innate hematoma resolution and neurological recovery after hemorrhagic stroke in mice. Proc. Natl Acad. Sci. U. S. A. 117, 32679–32690 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Frakes, A. E. et al. Microglia induce motor neuron death via the classical NF-κB pathway in amyotrophic lateral sclerosis. Neuron 81, 1009–1023 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Lavin, Y. et al. Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell 159, 1312–1326 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Reyna, M. A., Leiserson, M. D. M. & Raphael, B. J. Hierarchical HotNet: identifying hierarchies of altered subnetworks. Bioinformatics 34, i972–i980 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. 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 

  40. 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 

  41. 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 

  42. Du, Y. et al. LRRC25 inhibits type I IFN signaling by targeting ISG15-associated RIG-I for autophagic degradation. EMBO J. 37, 351–366 (2018).

    Article  CAS  PubMed  Google Scholar 

  43. Lee, M. et al. Acidic fibroblast growth factor (FGF) potentiates glial-mediated neurotoxicity by activating FGFR2 IIIb protein. J. Biol. Chem. 286, 41230–41245 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Pimenova, A. A. et al. Alzheimer’s-associated PU.1 expression levels regulate microglial inflammatory response. Neurobiol. Dis. 148, 105217 (2021).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  46. Wang, M. et al. The Mount Sinai cohort of large-scale genomic, transcriptomic and proteomic data in Alzheimer’s disease. Sci. Data 5, 180185 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Bennett, D. A., Schneider, J. A., Arvanitakis, Z. & Wilson, R. S. Overview and findings from the religious orders study. Curr. Alzheimer Res. 9, 628–645 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Bennett, D. A. et al. Overview and findings from the rush Memory and Aging Project. Curr. Alzheimer Res. 9, 646–663 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Buenrostro, J. D., Wu, B., Chang, H. Y. & Greenleaf, W. J. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 109, 21.29.1–21.29.9 (2015).

    Article  Google Scholar 

  50. Zhang, Y. et al. Model-based analysis of ChIP-seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    CAS  PubMed  Google Scholar 

  53. van de Geijn, B., McVicker, G., Gilad, Y. & Pritchard, J. K. WASP: allele-specific software for robust molecular quantitative trait locus discovery. Nat. Methods 12, 1061–1063 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinf. 12, 323 (2011).

    Article  CAS  Google Scholar 

  55. Lachmann, A. et al. Massive mining of publicly available RNA-seq data from human and mouse. Nat. Commun. 9, 1366 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Rao, S. S. P. et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 159, 1665–1680 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Servant, N. et al. HiC-Pro: an optimized and flexible pipeline for Hi-C data processing. Genome Biol. 16, 259 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Imakaev, M. et al. Iterative correction of Hi-C data reveals hallmarks of chromosome organization. Nat. Methods 9, 999–1003 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Weirauch, M. T. et al. Determination and inference of eukaryotic transcription factor sequence specificity. Cell 158, 1431–1443 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. 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 

  61. Sidak, Z. Rectangular confidence regions for the means of multivariate normal distributions. J. Am. Stat. Assoc. 62, 626 (1967).

    Google Scholar 

  62. Yoshida, H. et al. The cis-regulatory atlas of the mouse immune system. Cell 176, 897–912.e20 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Anscombe, F. J. The transformation of poisson, binomial and negative-binomial data. Biometrika 35, 246–254 (1948).

    Article  Google Scholar 

  64. Gilmour, A. R., Thompson, R. & Cullis, B. R. Average information REML: an efficient algorithm for variance parameter estimation in linear mixed models. Biometrics 51, 1440 (1995).

    Article  Google Scholar 

  65. Han, B. & Eskin, E. Random-effects model aimed at discovering associations in meta-analysis of genome-wide association studies. Am. J. Hum. Genet. 88, 586–598 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Coetzee, S. G., Coetzee, G. A. & Hazelett, D. J. motifbreakR: an R/Bioconductor package for predicting variant effects at transcription factor binding sites. Bioinformatics 31, 3847–3849 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. 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 

  69. International HapMap Consortium. The international hapmap project. Nature 426, 789–796 (2003).

    Article  CAS  Google Scholar 

  70. 1000 Genomes Project Consortium et al. A global reference for human genetic variation. Nature 526, 68–74 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the patients and families who donated material for these studies. We thank the computational resources and staff expertise provided by the Scientific Computing group at the Icahn School of Medicine at Mount Sinai. This study was supported by the National Institute on Aging, National Institutes of Health grants R01-AG050986 (P.R.), R01-AG067025 (P.R. and V.H.) and R01-AG065582 (P.R. and V.H.). J.B. was supported in part by Alzheimer’s Association Research Fellowship AARF-21-722200. P.D. is partially supported by NARSAD Young Investigator Grant 29683 from the Brain and Behavior Research Foundation.

Author information

Authors and Affiliations

Authors

Contributions

J.F.F., V.H. and P.R. conceived of and initiated the project. J.F.F. and P.R. designed experimental strategies for omics profiling. A.W.C., B.K., D.B, C.P.K. and V.H. provided human brain tissue dissections and biopsies. J.F.F., S.R., S.P.K. and Z.S. performed data generation. R.K., B.Z., J.B., G.E.H. and P.R. designed analytical strategies. R.K., J.B. and P.D. conducted initial bioinformatics, sample processing and QC for the omics data. R.K. and G.E.H. developed the computational scheme and performed the downstream analysis. B.Z. performed the QTL analysis. J.B. performed the TF analysis. P.D. performed the Hi-C analysis. K.G. performed ChIP analysis. J.H., K.P.L. and T.R. provided additional QTL resources. J.F.F. and P.R. supervised overall data generation. G.E.H. and P.R. supervised overall data analysis. R.K., J.F.F., B.Z., J.B., G.E.H. and P.R. wrote the manuscript with input from all authors.

Corresponding authors

Correspondence to Roman Kosoy, Gabriel E. Hoffman or Panos Roussos.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks Inge Holtman, Jeremy Schwartzentruber, 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 Schematic outline of data generation and integrative analyses.

The general schema of the data generation, processing and utilization in the analyses described.

Extended Data Fig. 2 FACS gating of fresh microglia.

The gating strategy for a representative sample targeting live (DAPI-) CD45+ microglia.

Extended Data Fig. 3 Comparison of human microglia RNA-seq dataset to other available microglia datasets.

Comparison of human microglia RNA-seq dataset to other available microglia datasets. We applied multidimensional scaling (MDS) to expression data processed by the standard RAPiD pipeline (https://github.com/CommonMindConsortium/RAPiD-nf/ blob/master/tutorial.md) for the two microglia transcriptomics datasets whose eQTLs were utilized in meta-eQTL analyses, as well as other relevant cell types. a) Comparisons include other brain-derived populations and b) only the three microglia and one monocyte datasets.

Extended Data Fig. 4 Comparison of caQTLs and eQTLs identified in our human microglia to relevant publicly available datasets.

a) π1 replication of caQTLs identified in our microglia data to macrophage caQTLs from Alasoo et al. 18 and brain homogenate from CommonMind 19; b) π1 replication of eQTLs identified in our microglia data to microglia eQTLs from Young et al. 7 and Lopes et al. 8, and to macrophage eQTLs from Alasoo et.al 18.

Extended Data Fig. 5 The distribution and LDsc analyses of colocalized eQTLs and caQTLs.

a) Venn diagrams represent the colocalized 1,457 OCR-Gene pairs among meta-caQTLs (green) and meta-eQTLs (red). Colocalized OCR-Gene pairs include 16 OCRs and 14 genes not significant in standalone meta-caQTLs and meta-eQTLs. b) The distribution of the distance between the OCRs and genes for the colocalized caQTLs and eQTL pairs. Mean distance is 101 kb, and median distance is 53 kb. c) Overlap between OCR-gene links identified via caQTL-eQTL colocalization method versus E-P links identified via the ABC approach. d) CaSNPs for the caQTLs colocalized with eQTLs are more likely to be located within OCRABC than in random OCRs. Red line represents the number of OCRABC regulated by colocalized caQTL, versus the distribution of OCRABC from 100 random sets of 1,457 distance-matched eQTL/caQTL pairs (P = 4.2 × 10-12, two-sided t-test). e) Enrichment of SNPs representing 95% confidence intervals for the meta-caQTL and meta-eQTL sets identified in fresh microglia via LDscore analyses using summary statistics from a set of selected GWAS studies. For each genetic variant the value utilized was the highest product between posterior probabilities from eQTL and caQTL analyses (n = 30,028). Values in dark blue represent nominally significant enrichment. The utilized GWAS studies are described in Supplementary Table 6, including the number of snps from each study. Coefficients from LD score regression (two-sided linear regression) are normalized by the per-SNP heritability (h2 /total SNPs per GWAS), with horizontal bars indicating SE (AD P = 0.036, FDR P = 0.17).

Extended Data Fig. 6 Integration of the landscape of AD etiology with genetic regulation of transcriptional and chromatin accessibility in microglia within the EPHA1-AS1 locus.

a) Local plot showing results from AD GWAS 13, eQTL analysis of EPHA1/EPHA1-AS1, and caQTL analysis of peak_188003 (P values are nominal significance estimates from two-sided logistic regression for GWAS, and mixed linear regression for QTL analyses). CLPP is the joint posterior probabilities 47 between the eSNP/caSNP PP and Jansen AD GWAS PP. Red points indicate genetic variants in the 95% credible set from statistical fine-mapping of each trait. Inset shows colocalization posterior probabilities (CLPP) for the top variant in the credible set for gene expression and chromatin accessibility. b) Visualization of EPHA1/EPHA1-AS1 locus showing open chromatin regions from four cell populations five and microglia from this study, E-P interactions (ABC), fine-mapped variants from AD (Jansen) 13, genetic regulation from eQTLs and caQTL from this study and colocalization analysis between pairs of traits (that is, AD GWAS, gene expression chromatin accessibility) using ‘coloc’ and all three traits using ‘moloc’ methods with Jansen et al. AD GWAS (PP15 > 0.5) 29.

Extended Data Fig. 7 Relationship between colocalized components and predicted functional annotation at EPHA1-AS1 locus.

Relationship between EPHA1-AS1 expression, OCR peak_188003 and rs11771145. a) Application of Causal Inference Test 48 identifies a genetic variant’s regulation on transcriptional activity of AD-implicated genes mediated by its effect on chromatin accessibility (one-sided Omnibus F-statistics). b) Correlation between the expression of EPHA1 and EPHA1-AS1, ATAC-seq signal at OCR peak_188003 and the genotype of rs11771145 (two-sided Spearman test). c) Predicted function of EPHA1-AS1 based on coexpression structure. lncHUB 31 predicts gene set annotations of every gene based on genome-wide co-expression structure and known annotations. Results are from query https://maayanlab.cloud/lnchub/?lnc=EPHA1-AS1.

Extended Data Fig. 8 Local manhattan plot of AD GWAS 13 and eQTL analysis for FIBP.

Red points indicate variants within the 95% confidence interval from statistical fine-mapping. P values are nominal significance estimates from two-sided logistic regression for GWAS and mixed linear regression for QTL analyses.

Extended Data Fig. 9 Local manhattan plot of AD GWAS 13 and eQTL analysis for LRRC25.

Red points indicate variants within the 95% confidence interval from statistical fine-mapping. P values are nominal significance estimates from two-sided logistic regression for GWAS and mixed linear regression for QTL analyses.

Extended Data Fig. 10 Local manhattan plot of AD GWAS 13, eQTL analysis for KCNN4 and caQTL for Peak_82668.

Bottom row indicates open chromatin regions in the window, and the red region indicates the target peak for caQTL analysis shown. Red points indicate variants within the 95% confidence interval from statistical fine-mapping. P values are nominal significance estimates from two-sided logistic regression for GWAS and mixed linear regression for QTL analyses.

Supplementary information

Supplementary Information

Extended Methods, Supplementary Results and Discussion and Supplementary Figures 1–19.

Reporting Summary

Peer Review File

Supplementary Table 1

Supplementary Tables 1–19.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kosoy, R., Fullard, J.F., Zeng, B. et al. Genetics of the human microglia regulome refines Alzheimer’s disease risk loci. Nat Genet 54, 1145–1154 (2022). https://doi.org/10.1038/s41588-022-01149-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-022-01149-1

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research