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:

DNA methylation QTL mapping across diverse human tissues provides molecular links between genetic variation and complex traits

Abstract

Studies of DNA methylation (DNAm) in solid human tissues are relatively scarce; tissue-specific characterization of DNAm is needed to understand its role in gene regulation and its relevance to complex traits. We generated array-based DNAm profiles for 987 human samples from the Genotype-Tissue Expression (GTEx) project, representing 9 tissue types and 424 subjects. We characterized methylome and transcriptome correlations (eQTMs), genetic regulation in cis (mQTLs and eQTLs) across tissues and e/mQTLs links to complex traits. We identified mQTLs for 286,152 CpG sites, many of which (>5%) show tissue specificity, and mQTL colocalizations with 2,254 distinct GWAS hits across 83 traits. For 91% of these loci, a candidate gene link was identified by integration of functional maps, including eQTMs, and/or eQTL colocalization, but only 33% of loci involved an eQTL and mQTL present in the same tissue type. With this DNAm-focused integrative analysis, we contribute to the understanding of molecular regulatory mechanisms in human tissues and their impact on complex traits.

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: Scheme of data generation and analysis overview.
Fig. 2: mQTL discovery and e/mQTL functional mechanism characterization.
Fig. 3: Characterization of HOXD-locus e/mQTL pleiotropy in the ovary.
Fig. 4: Colocalization of mQTLs and eQTLs with GWAS traits.
Fig. 5: Examples of trait-linked e/mQTLs.
Fig. 6: Characteristics of trait-linked mQTLs.

Similar content being viewed by others

Data availability

Summary statistics of mQTLs are available at the GTEx Portal (https://gtexportal.org/home/datasets). DNAm normalized data is available at GEO (GSE213478). All GTEx protected data are available via dbGaP (phs000424.v9); access to the DNAm raw data is provided through the AnVIL platform (https://anvil.terra.bio/#workspaces/anvil-datastorage/AnVIL_GTEx_V9_hg38). Independent linkage disequilibrium blocks coordinates to define GWAS hit loci, colocalization summary statistics and priors, single-tissue functional annotation enrichment statistics, and data to generate figures, are available at Figshare (https://figshare.com/projects/DNA_methylation_QTL_mapping_across_diverse_human_tissues_provides_molecular_links_between_genetic_variation_and_complex_traits/149524).

Code availability

Code for QTL and eQTM mapping, functional enrichment, and colocalization, as well as code to to generate manuscript figures, is available at the github repository (https://github.com/meritxellop/eGTEx_mQTLs_eQTLs_GWAS) and archived at zenodo (https://doi.org/10.5281/zenodo.7106660)99.

References

  1. Nicolae, D. L. et al. Trait-associated SNPs are more likely to be eQTLs: annotation to enhance discovery from GWAS. PLoS Genet. 6, e1000888 (2010).

    Article  Google Scholar 

  2. Cano-Gamez, E. & Trynka, G. From GWAS to function: Using functional genomics to identify the mechanisms underlying complex diseases. Front. Genet. 11, 424 (2020).

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

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

    Article  Google Scholar 

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

    Article  Google Scholar 

  6. Ongen, H. et al. Estimating the causal tissues for complex traits and diseases. Nat. Genet. 49, 1676–1683 (2017).

    Article  CAS  Google Scholar 

  7. Gamazon, E. R. et al. Using an atlas of gene regulation across 44 human tissues to inform complex disease- and trait-associated variation. Nat. Genet. 50, 956–967 (2018).

    Article  CAS  Google Scholar 

  8. Banovich, N. E. et al. Methylation QTLs are associated with coordinated changes in transcription factor binding, histone modifications, and gene expression levels. PLoS Genet. 10, e1004663 (2014).

    Article  Google Scholar 

  9. Li, E., Beard, C. & Jaenisch, R. Role for DNA methylation in genomic imprinting. Nature 366, 362–365 (1993).

    Article  CAS  Google Scholar 

  10. Payer, B. & Lee, J. T. X chromosome dosage compensation: how mammals keep the balance. Annu. Rev. Genet. 42, 733–772 (2008).

    Article  CAS  Google Scholar 

  11. Maurano, M. T. et al. Role of DNA methylation in modulating transcription factor occupancy. Cell Rep. 12, 1184–1195 (2015).

    Article  CAS  Google Scholar 

  12. Jin, Z. & Liu, Y. DNA methylation in human diseases. Genes Dis. 5, 1–8 (2018).

    Article  CAS  Google Scholar 

  13. Kaminsky, Z. A. et al. DNA methylation profiles in monozygotic and dizygotic twins. Nat. Genet. 41, 240–245 (2009).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  15. van Dongen, J. et al. Genetic and environmental influences interact with age and sex in shaping the human methylome. Nat. Commun. 7, 11115 (2016). vol.

    Article  Google Scholar 

  16. Cheung, W. A. et al. Functional variation in allelic methylomes underscores a strong genetic contribution and reveals novel epigenetic alterations in the human epigenome. Genome Biol. 18, 1–21 (2017).

    Article  Google Scholar 

  17. Volkov, P. et al. A genome-wide mQTL analysis in human adipose tissue identifies genetic variants associated with DNA methylation, gene expression and metabolic traits. PLoS One 11, e0157776 (2016).

    Article  Google Scholar 

  18. Hannon, E. et al. Methylation QTLs in the developing brain and their enrichment in schizophrenia risk loci. Nat. Neurosci. 19, 48–54 (2016).

    Article  CAS  Google Scholar 

  19. Morrow, J. D. et al. Human lung DNA methylation quantitative trait loci colocalize with chronic obstructive pulmonary disease genome-wide association loci. Am. J. Respir. Crit. Care Med. 197, 1275–1284 (2018).

    Article  CAS  Google Scholar 

  20. Taylor, D. L. et al. Integrative analysis of gene expression, DNA methylation, physiological traits, and genetic variation in human skeletal muscle. Proc. Natl Acad. Sci. U. S. A. 116, 10883–10888 (2019).

    Article  CAS  Google Scholar 

  21. Huan, T. et al. Genome-wide identification of DNA methylation QTLs in whole blood highlights pathways for cardiovascular disease. Nat. Commun. 10, 4267 (2019).

    Article  Google Scholar 

  22. Andrews, S. V. et al. Cross-tissue integration of genetic and epigenetic data offers insight into autism spectrum disorder. Nat. Commun. 8, 1–10 (2017).

    Article  CAS  Google Scholar 

  23. 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  Google Scholar 

  24. Gibbs, J. R. et al. Abundant quantitative trait loci exist for DNA methylation and gene expression in human brain. PLoS Genet. 6, e1000952 (2010).

    Article  Google Scholar 

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

    Article  Google Scholar 

  26. Gutierrez-Arcelus, M. et al. Tissue-specific effects of genetic and epigenetic variation on gene regulation and splicing. PLoS Genet. 11, e1004958 (2015).

    Article  Google Scholar 

  27. Schulz, H. et al. Genome-wide mapping of genetic determinants influencing DNA methylation and gene expression in human hippocampus. Nat. Commun. 8, 1511 (2017). vol.

    Article  Google Scholar 

  28. Do, C. et al. Mechanisms and disease associations of haplotype-dependent allele-specific DNA methylation. Am. J. Hum. Genet. 98, 934–955 (2016).

    Article  CAS  Google Scholar 

  29. Grundberg, E. et al. Global analysis of DNA methylation variation in adipose tissue from twins reveals links to disease-associated variants in distal regulatory elements. Am. J. Hum. Genet. 93, 876–890 (2013). 11/.

    Article  CAS  Google Scholar 

  30. Bell, J. T. et al. DNA methylation patterns associate with genetic and gene expression variation in HapMap cell lines. Genome Biol. 12, R10 (2011).

    Article  CAS  Google Scholar 

  31. McClay, J. L. et al. High density methylation QTL analysis in human blood via next-generation sequencing of the methylated genomic DNA fraction. Genome Biol. 16, 291 (2015).

    Article  Google Scholar 

  32. Pierce, B. L. et al. Co-occurring expression and methylation QTLs allow detection of common causal variants and shared biological mechanisms. Nat. Commun. 9, 804 (2018).

    Article  Google Scholar 

  33. Min, J. L. et al. Genomic and phenotypic insights from an atlas of genetic effects on DNA methylation. Nat. Genet. 53, 1311–1321 (2021).

    Article  CAS  Google Scholar 

  34. Bonder, M. J. et al. Disease variants alter transcription factor levels and methylation of their binding sites. Nat. Genet. 49, 131 (2017).

    Article  CAS  Google Scholar 

  35. Qi, T. et al. Identifying gene targets for brain-related traits using transcriptomic and methylomic data from blood. Nat. Commun. 9, 1–12 (2018).

    Article  Google Scholar 

  36. Schultz, M. D. et al. Human body epigenome maps reveal noncanonical DNA methylation variation. Nature 523, 212–216 (2015).

    Article  CAS  Google Scholar 

  37. Kerimov, N. et al. A compendium of uniformly processed human gene expression and splicing quantitative trait loci. Nat. Genet. 53, 1290–1299 (2021).

    Article  CAS  Google Scholar 

  38. Zheng, Z. et al. QTLbase: an integrative resource for quantitative trait loci across multiple human molecular phenotypes. Nucleic Acids Res. 48, D983–D991 (2020).

    Article  CAS  Google Scholar 

  39. eGTEx Project. Enhancing GTEx by bridging the gaps between genotype, gene expression, and disease. Nat. Genet. 49, 1664–1670 (2017).

    Article  Google Scholar 

  40. Kim, S. et al. Expression quantitative trait methylation analysis reveals methylomic associations with gene expression in childhood asthma. Chest 158, 1841–1856 (2020).

    Article  CAS  Google Scholar 

  41. Bommarito, P. A. & Fry, R. C. The role of DNA methylation in gene regulation, in Toxicoepigenetics (eds. McCullough, S. D. & Dolinoy, D. C.) 127–151 (Academic Press, 2019).

  42. Urbut, S. M., Wang, G., Carbonetto, P. & Stephens, M. Flexible statistical methods for estimating and testing effects in genomic studies with multiple conditions. Nat. Genet. 51, 187–195 (2019).

    Article  CAS  Google Scholar 

  43. Brown, A. A. et al. Predicting causal variants affecting expression by using whole-genome sequencing and RNA-seq from multiple human tissues. Nat. Genet. 49, 1747–1751 (2017).

    Article  CAS  Google Scholar 

  44. Perzel Mandell, K. A. et al. Genome-wide sequencing-based identification of methylation quantitative trait loci and their role in schizophrenia risk. Nat. Commun. 12, 5251 (2021).

    Article  CAS  Google Scholar 

  45. Ziller, M. J. et al. Charting a dynamic DNA methylation landscape of the human genome. Nature 500, 477–481 (2013).

    Article  CAS  Google Scholar 

  46. Schübeler, D. Function and information content of DNA methylation. Nature 517, 321–326 (2015).

    Article  Google Scholar 

  47. Bell, C. G. The epigenomic analysis of human obesity. Obesity 25, 1471–1481 (2017).

    Article  CAS  Google Scholar 

  48. Villicaña, S. & Bell, J. T. Genetic impacts on DNA methylation: research findings and future perspectives. Genome Biol. 22, 127 (2021).

    Article  Google Scholar 

  49. Pai, A. A., Pritchard, J. K. & Gilad, Y. The genetic and mechanistic basis for variation in gene regulation. PLoS Genet. 11, e1004857 (2015).

    Article  Google Scholar 

  50. Wang, M. et al. Identification of DNA motifs that regulate DNA methylation. Nucleic Acids Res. 47, 6753–6768 (2019).

    Article  CAS  Google Scholar 

  51. Wu, Y. et al. Integrative analysis of omics summary data reveals putative mechanisms underlying complex traits. Nat. Commun. 9, 918 (2018).

    Article  Google Scholar 

  52. Zhang, W., Spector, T. D., Deloukas, P., Bell, J. T. & Engelhardt, B. E. Predicting genome-wide DNA methylation using methylation marks, genomic position, and DNA regulatory elements. Genome Biol. 16, 14 (2015).

    Article  Google Scholar 

  53. Liu, Y. et al. GeMes, clusters of DNA methylation under genetic control, can inform genetic and epigenetic analysis of disease. Am. J. Hum. Genet. 94, 485–495 (2014).

    Article  CAS  Google Scholar 

  54. Goode, E. L. et al. A genome-wide association study identifies susceptibility loci for ovarian cancer at 2q31 and 8q24. Nat. Genet. 42, 874–879 (2010).

    Article  CAS  Google Scholar 

  55. Kar, S. P. et al. Network-based integration of GWAS and gene expression identifies a HOX-centric network associated with serous ovarian cancer risk. Cancer Epidemiol. Biomark. Prev. 24, 1574–1584 (2015).

    Article  CAS  Google Scholar 

  56. Shah, N. & Sukumar, S. The Hox genes and their roles in oncogenesis. Nat. Rev. Cancer 10, 361–371 (2010).

    Article  CAS  Google Scholar 

  57. Zhao, T., Hu, Y., Zang, T. & Wang, Y. Integrate GWAS, eQTL, and mQTL data to identify Alzheimer’s disease-related genes. Front. Genet. 10, 1021 (2019).

    Article  CAS  Google Scholar 

  58. Soliai, M. M. et al. Multi-omics colocalization with genome-wide association studies reveals a context-specific genetic mechanism at a childhood onset asthma risk locus. Genome Med. 13, 157 (2021).

    Article  CAS  Google Scholar 

  59. Bojesen, S. E. et al. Multiple independent variants at the TERT locus are associated with telomere length and risks of breast and ovarian cancer. Nat. Genet. 45, 371–384 (2013). 384e1–2.

    Article  CAS  Google Scholar 

  60. Huan, T. et al. Genome-wide identification of microRNA expression quantitative trait loci. Nat. Commun. 6, 6601 (2015).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  62. Gleason, K. J., Yang, F., Pierce, B. L., He, X. & Chen, L. S. Primo: integration of multiple GWAS and omics QTL summary statistics for elucidation of molecular mechanisms of trait-associated SNPs and detection of pleiotropy in complex traits. Genome Biol. 21, 236 (2020).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  64. Fishilevich, S. et al. GeneHancer: genome-wide integration of enhancers and target genes in GeneCards. Database 2017, bax028 (2017).

    Article  Google Scholar 

  65. Eales, J. M. et al. Uncovering genetic mechanisms of hypertension through multi-omic analysis of the kidney. Nat. Genet. 53, 630–637 (2021).

    Article  CAS  Google Scholar 

  66. Ghoussaini, M. et al. Open Targets Genetics: systematic identification of trait-associated genes using large-scale genetics and functional genomics. Nucleic Acids Res. 49, D1311–D1320 (2021).

    Article  CAS  Google Scholar 

  67. Maunakea, A. K., Chepelev, I., Cui, K. & Zhao, K. Intragenic DNA methylation modulates alternative splicing by recruiting MeCP2 to promote exon recognition. Cell Res. 23, 1256–1269 (2013).

    Article  CAS  Google Scholar 

  68. Umans, B. D., Battle, A. & Gilad, Y. Where are the disease-associated eQTLs? Trends Genet. 37, 109–124 (2021).

    Article  CAS  Google Scholar 

  69. Kapoor, M. et al. Multi-omics integration analysis identifies novel genes for alcoholism with potential overlap with neurodegenerative diseases. Nat. Commun. 12, 5071 (2021).

    Article  CAS  Google Scholar 

  70. Hemani, G., Bowden, J. & Davey Smith, G. Evaluating the potential role of pleiotropy in Mendelian randomization studies. Hum. Mol. Genet. 27, R195–R208 (2018).

    Article  CAS  Google Scholar 

  71. Niemöller, C. et al. Bisulfite-free epigenomics and genomics of single cells through methylation-sensitive restriction. Commun. Biol. 4, 153 (2021).

    Article  Google Scholar 

  72. Nuñez, J. K. et al. Genome-wide programmable transcriptional memory by CRISPR-based epigenome editing. Cell 184, 2503–2519.e17 (2021).

    Article  Google Scholar 

  73. Hawe, J. S. et al. Genetic variation influencing DNA methylation provides insights into molecular mechanisms regulating genomic function. Nat. Genet. 54, 18–29 (2022).

    Article  CAS  Google Scholar 

  74. Jiang, L. et al. A quantitative proteome map of the human body. Cell 83, 269–283.e19 (2020).

    Article  Google Scholar 

  75. Rizzardi, L. F. et al. Human brain region-specific variably methylated regions are enriched for heritability of distinct neuropsychiatric traits. Genome Biol. 22, 116 (2021).

    Article  CAS  Google Scholar 

  76. Siminoff, L. A., Wilson-Genderson, M., Gardiner, H. M., Mosavel, M. & Barker, K. L. Consent to a postmortem tissue procurement study: Distinguishing family decision makers’ knowledge of the Genotype-Tissue Expression Project. Biopreserv. Biobank. 16, 200–206 (2018).

    Article  Google Scholar 

  77. Ali, T., Renkawitz, R. & Bartkuhn, M. Insulators and domains of gene expression. Curr. Opin. Genet. Dev. 37, 17–26 (2016).

    Article  CAS  Google Scholar 

  78. Morris, T. J. et al. ChAMP: 450k Chip Analysis Methylation Pipeline. Bioinformatics 30, 428–430 (2014).

    Article  CAS  Google Scholar 

  79. Pidsley, R. et al. Critical evaluation of the Illumina MethylationEPIC BeadChip microarray for whole-genome DNA methylation profiling. Genome Biol. 17, 208 (2016).

    Article  Google Scholar 

  80. Fortin, J.-P., Triche, T. J. Jr & Hansen, K. D. Preprocessing, normalization and integration of the Illumina HumanMethylationEPIC array with minfi. Bioinformatics 33, 558–560 (2017).

    CAS  Google Scholar 

  81. Triche, T. J. Jr, Weisenberger, D. J., Van Den Berg, D., Laird, P. W. & Siegmund, K. D. Low-level processing of Illumina Infinium DNA Methylation BeadArrays. Nucleic Acids Res. 41, e90 (2013).

    Article  CAS  Google Scholar 

  82. Teschendorff, A. E. et al. A beta-mixture quantile normalization method for correcting probe design bias in Illumina Infinium 450 k DNA methylation data. Bioinformatics 29, 189–196 (2013).

    Article  CAS  Google Scholar 

  83. Suzuki, R. & Shimodaira, H. pvclust: Hierarchical Clustering with P-Values via Multiscale Bootstrap Resampling. https://CRAN.R-project.org/package=pvclust (2015).

  84. Stegle, O., Parts, L., Piipari, M., Winn, J. & Durbin, R. Using probabilistic estimation of expression residuals (PEER) to obtain increased power and interpretability of gene expression analyses. Nat. Protoc. 7, 500–507 (2012).

    Article  CAS  Google Scholar 

  85. Leek, J. T. & Storey, J. D. Capturing heterogeneity in gene expression studies by surrogate variable analysis. PLoS Genet. 3, 1724–1735 (2007).

    Article  CAS  Google Scholar 

  86. Storey, J. D. & Tibshirani, R. Statistical significance for genomewide studies. Proc. Natl Acad. Sci. U. S. A. 100, 9440–9445 (2003).

    Article  CAS  Google Scholar 

  87. Ongen, H., Buil, A., Brown, A. A., Dermitzakis, E. T. & Delaneau, O. Fast and efficient QTL mapper for thousands of molecular phenotypes. Bioinformatics 32, 1479–1485 (2016).

    Article  CAS  Google Scholar 

  88. Wen, X. Molecular QTL discovery incorporating genomic annotations using Bayesian false discovery rate control. Ann. Appl. Stat. 10, 1619–1638 (2016).

    Article  Google Scholar 

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

    Article  Google Scholar 

  90. Phelan, C. M. et al. Identification of 12 new susceptibility loci for different histotypes of epithelial ovarian cancer. Nat. Genet. 49, 680–691 (2017).

    Article  CAS  Google Scholar 

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

    CAS  Google Scholar 

  92. Wen, X., Pique-Regi, R. & Luca, F. Integrating molecular QTL data into genome-wide genetic association analysis: Probabilistic assessment of enrichment and colocalization. PLoS Genet. 13, e1006646 (2017).

    Article  Google Scholar 

  93. Pividori, M. et al. PhenomeXcan: Mapping the genome to the phenome through the transcriptome. Sci. Adv. 6 (2020).

  94. Hukku, A. et al. Probabilistic colocalization of genetic variants from complex and molecular traits: promise and limitations. Am. J. Hum. Genet. 108, 25–35 (2021).

    Article  CAS  Google Scholar 

  95. Wen, X., Lee, Y., Luca, F. & Pique-Regi, R. Efficient integrative multi-SNP association analysis via deterministic approximation of posteriors. Am. J. Hum. Genet. 98, 1114–1129 (2016).

    Article  CAS  Google Scholar 

  96. Foley, C. N. et al. A fast and efficient colocalization algorithm for identifying shared genetic risk factors across multiple traits. Nat. Commun. 12, 764 (2021).

    Article  CAS  Google Scholar 

  97. Võsa, U. et al. Large-scale cis- and trans-eQTL analyses identify thousands of genetic loci and polygenic scores that regulate blood gene expression.Nat. Genet. 53, 1300–1310 (2021).

    Article  Google Scholar 

  98. Bonder, M. J. et al. Identification of rare and common regulatory variants in pluripotent cells using population-scale transcriptomics. Nat. Genet. 53, 313–321 (2021).

    Article  CAS  Google Scholar 

  99. Oliva, M. eGTEx_mQTLs_eQTLs_GWAS: DNA methylation QTL mapping across diverse human tissues provides molecular links between genetic variation and complex traits. Code resource. GitHub: https://github.com/meritxellop/eGTEx_mQTLs_eQTLs_GWAS; Zenodo: https://doi.org/10.5281/zenodo.7106660

Download references

Acknowledgements

This work was supported by grants U01 HG007601 (to B.L.P.), R35ES028379 (to B.L.P.), 2R01 GM108711 (to L.S.C) and U24 CA210993-SUB (to L.S.C) and was completed in part with computational resources provided by the Center for Research Informatics at the University of Chicago. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. We thank the donors and their families for their generous gifts of biospecimens to the GTEx research project; the Genomics Platform at the Broad Institute for data generation; F. Aguet, J. Nedzel and K. Ardlie for sample delivery logistics and data release management; D. Delgado and L. Tong for assistance with assessing mQTL replication; and M. Goraj, J. Witkos, J. Degner and J. Resztak for comments on an earlier version of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

B.L.P. and M.O. conceived the study; M.O. conceived and led all analysis supervised by B.L.P. and L.S.C.; M.O. performed all bioinformatic analysis, granted K.D., M.C. and Y.L. contributions; M.O. led the writing and editing of the manuscript and supplement; B.L.P., L.S.C. and H.A. contributed to the editing of the manuscript and supplement; M.O., B.L.P. and L.S.C. coordinated analyses of all contributing authors; F.J. generated the DNAm data; M.G.K. supervised the generation of the DNAm data; K.D. processed and QC-ed the DNAm data; Y.L. and M.C. contributed to the mQTL functional characterization analysis. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Meritxell Oliva, Lin S. Chen or Brandon L. Pierce.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks the anonymous reviewers for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Characterization of methylomes across tissues, eQTM discovery and tissue specificity patterns.

(a) Sample similarity based on DNAm profiles. Dimensionality reduction was performed with a t-Distributed Stochastic Neighbor Embedding approach (t-SNE). (b) Hierarchical tissue clustering based on complete methylomes (left panel) and transcriptomes (right panel) of nine tissues (x axis). The molecular phenotypes displayed (y axis) correspond to the top 20,000 most divergent CpG sites and genes across tissues. DNAm and gene expression values are column-wise scaled. (c) Number of eQTMs per tissue, defined at LFSR < 0.05 or FDR < 0.05, shown with per-tissue eQTM-mapping sample sizes in parentheses. FDR: False Discovery Rate. LFSR: Local False Sign Rate. (d) Tissue sharing profile of eQTMs. (e) Contribution (x axis, square-root transformed log(OR)) of selected factors to eQTM likelihood (presence) for different gene regulatory elements (y axis). Dist.: Distance. OR: Odds Ratio. Factor units: CpG–gene distance [Kb], eQTM Sign [‘1’ for negative correlation between methylation and expression, ‘0’ otherwise], CpG DNAm [M-value], gene expression [log2(TPM + 1)]. OR estimates were derived from across-tissue meta-analysis (nine tissues) of predictor coefficients of eQTM likelihood, fitted with a logistic regression model (Methods).

Extended Data Fig. 2 DNAm-derived PEER factors association with technical, clinical and epidemiological covariates.

Proportion of variance - mean adjusted R2 across top three PEERs (R2adj) - of the PEER factors explained in part by known donor and sample clinical and biological covariates. Each cell shows the proportion of variance explained by the covariate with respect to the three top PEERs in a specific tissue. Only covariates with R2adj ≥ 0.02 in any tissue are shown. Tissues and covariates are ordered based on hierarchical clustering with complete agglomeration with Euclidean distance. Gray cells indicate unavailable data. The cells at the bottom of the panel shows that the PEERs are capturing batch effects, as expected.

Extended Data Fig. 3 DNAm-derived PEER factors association with tissue cellular abundances.

Proportion of variance - mean adjusted R2 across top three PEERs (R2adj) - of the PEER factors explained in part by tissue cellular abundances. Each bar shows the proportion of variance explained by the cell abundance with respect to the three top PEERs in a specific tissue. (b) Fraction of cell abundance (y axis) estimated by DNAm cell-type deconvolution with EpiSCORE, stratified by cell type (x axis) in corresponding tissue. Breast cell abundances are stratified by sex to illustrate sex-differential cell abundances. Cell abundances were estimated for all available samples per tissue: NBreast,Males = 14, NBreast,Females = 38, NBlood,Pooled = 54, NColon,Pooled = 224. B: B cells, NK: Natural Killer cells, CD4T: CD4+ T-cells, CD8T: CD8+ T-cells, Mono: Monocytes, Neutro: Neutrophils, Eosino: Eosinophils, Basal: Basal Epithelial cells, EC: Endothelial cells, Fat: Adipocytes, Luminal: Luminal Epithelial cells, Lym: Lymphocytes, Macro: Macrophages, Epi: Epithelial cells, Mye: Myeloid cells, Stromal: Stromal cells.

Extended Data Fig. 4 Tissue specificity of QTLs.

(a) Tissue sharing profile of mQTLs and eQTLs. (b-c) Cross-tissue sharing of mCpGs and eGenes. Cross-tissue mean percent of mCpGs and eGenes per tissue-sharing category is shown in parentheses. Of note, testis is an outlier for eQTL tissue specificity, as 23.5% of eGenes were not detected in any other tissue. Avg.: average (mean). (c) Cross-tissue sharing of mQTL tissue-leveraged effect magnitudes (y axis) per gene regulatory region (x axis, 36 data points per box plot). P-values of paired two-sided Wilcoxon signed rank tests are shown for corresponding pairwise comparisons; p-value of Kruskal-Wallis rank sum test is shown for the three-way comparison. Enh.: Enhancer. (d, e) Validation of tissue-specific mQTLs in muscle, blood and brain mQTL external cohorts, see (b) for tissue color legend. In (d), for each cohort, the average of absolute mQTL effect sizes and corresponding standard error is displayed for each set of tissue-specific mQTLs identified in each GTEx tissue (x axis). In (e), Spearman correlation between external and GTEx mQTL effect sizes, and associated standard error, is shown. Fisher’s z transformation was applied to Spearman correlation coefficients; standard errors were calculated based on the transformed coefficients. In (d,e), the number of QTL associations (N) tested for each pairwise comparison is as follows: NFUSION,GTEx = 195|4142|336,4630,287,4643|1428|360|369, NGoDMC,GTEx = 22|1353|47|1478|70|1019|292|83|102 and NROSMAP,GTEx = 57|2428|156|2587|163|2673|791|214|109, where GTEx corresponds to Breast|Colon|Kidney|Lung|Muscle|Ovary|Prostate|Testis|Blood tissues, respectively.

Extended Data Fig. 5 Representativity of GTEx mCpGs in external cohorts.

Overlap of mCpGs identified in GTEx (FDR < 0.05) with mCpGs identified in external mQTL cohorts at different nominal p-value thresholds (P < 1e-03 and P < 1e-05). Results are represented for all mCpGs - detected in EPIC and/or 450 K Illumina array - and for 450 K Illumina array CpG sites exclusively. P-value thresholds correspond to external cohort nominal mQTL associations, derived from QTL mapping by multiple regression two-sided t-tests.

Extended Data Fig. 6 Enrichment of QTLs in chromatin states.

(a) QTL enrichment (x-axis) in tissue-matching open chromatin regions derived from ENCODE DNase-seq profiles per tissue (y-axis). Whole blood is excluded due to lack of a tissue-matching DNase-seq profile. Enrichment differences between tissues may be due in part to per-tissue DNase-seq data quality. (b) QTL enrichment (x-axis) in active chromatin states. OR: Odds Ratio. Enrichment values correspond to maximum-likelihood estimated log(ORs) for single-tissue in (a) and from across-tissue (nine tissues) meta-analysis in (b). In all panels, whiskers represent the 95% confidence interval of the enrichment value.

Extended Data Fig. 7 Characterization of mQTL pleiotropy.

(a) Scheme of possible scenarios of eQTL-mQTL colocalization regarding QTL variants’ pleiotropic effect on multiple mCpGs and eGenes. (b) Quantification of mQTL-eQTL pleiotropy per tier per tissue, in percent of mCpGs belonging to each tier. Tier details illustrated in (a). Avg.: average (mean). (c) Distribution of the number of eGenes per mCpG (left panel) and mCpGs per eGene (right panel) involved in mQTL-eQTL colocalization events, stratified by tissue. Avg.: average (mean).

Extended Data Fig. 8 Evaluation of mQTL-GWAS colocalization approach.

(a) Density plot of mQTL-GWAS colocalization scores based on coloc run with default (y axis) and fastenloc-derived priors (x axis) on UKB standing height GWAS; Spearman’s rho is shown. Each dot corresponds to a colocalization test for a particular GWAS hit, independent mQTL and tissue combination. (b) Density plot of mQTL-GWAS colocalization scores based on coloc (x axis) and fastenloc (y axis) approaches on all GWASs; Spearman’s rho is shown. Each dot corresponds to a colocalization test for a particular GWAS, GWAS hit, independent mQTL and tissue combination. Dots within the top-right quadrant correspond to significant (RCP > 0.3 and PP4 > 0.3) colocalizations. PP4: coloc-derived posterior probability where the two traits share a single causal variant. RCP: fastenloc-derived probability of a genomic region harboring a colocalized signal.

Extended Data Fig. 9 Signatures of QTL-GWAS colocalizations and trait-linked QTLs.

(a) Percent of QTL-colocalized GWAS hits (y axis) per GWAS trait (x axis) stratified by GWAS trait category and colocalization group (see Fig. 4). Only GWAS traits with > = 10 colocalized GWAS hits are displayed. (b) Mean DNAm - in M-values - of mCpGs (left panel) and gene expression - in log2(TPM + 1) - of eGenes (right panel) tested for colocalization, stratified by tissue and colocalization group (see Fig. 4). Mean DNAm and gene expression across tissues is indicated by a dashed line. Whiskers represent the 95% confidence interval of the mean, calculated based on 5,000 replications of bootstrapped samples (random sampling with replacement). The number of mCpGs/eGenes (N) tested per bootstrap is as follows: NMeanMethylation = (12472|51|124), (147806|306|1111), (17574|24|221), (157356|359|1234), (13623|45|162), (127008|147|1041), (65147|103|60), (13576|38|101), (20127|126|254) and NMeanGeneExpression = (10050|27|168), (10800|110|103), (1147|4|22), (13053|144|149), (12594|33|218), (5120|55|47), (6744|43|75), (17025|18|164), (11545|95|291) for QTL-GWAS tested, e/mQTL-shared and e/mQTL-specific eGenes/mCpGs in Breast|Colon|Kidney|Lung|Muscle|Ovary|Prostate|Testis|Blood tissues, respectively.

Extended Data Fig. 10 Breast cancer linked e/mQTLs in the TERT-CLPTM1L locus.

Colocalized molecular phenotypes for this locus, identified by breast cancer GWAS-QTL multivariate colocalization approach (Methods), are provided in the top summary table. The mQTLs colocalizing with these breast cancer GWAS signals (that is, mCpGs cg03935379 and cg07380026) are shown in Fig. 5a, b. Additional details are provided in Supplementary Table 8. Plots illustrate association p-values in the locus for breast cancer estrogen positive (ER+) GWAS (top panel), breast cancer estrogen negative (ER−) GWAS (middle panel) and TERT eQTL signal in induced pluripotent stem cells (iPSCs) (bottom panel). Genotype-phenotype association p-values correspond to rs10069690, lead signal for breast cancer ER- GWAS. Linkage disequilibrium between loci is quantified by squared Pearson coefficient of correlation (r2) in population from European origin. Breast GWAS p-values were obtained from Milne et al. 2017, and iPSC TERT eQTL p-values from Bonder et al 2021. Mb: mega base. P-values correspond to nominal GWAS or QTL associations, derived from multiple regression two-sided t-tests.

Supplementary information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Oliva, M., Demanelis, K., Lu, Y. et al. DNA methylation QTL mapping across diverse human tissues provides molecular links between genetic variation and complex traits. Nat Genet 55, 112–122 (2023). https://doi.org/10.1038/s41588-022-01248-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-022-01248-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