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:

Allelic imbalance of chromatin accessibility in cancer identifies candidate causal risk variants and their mechanisms

Abstract

While many germline cancer risk variants have been identified through genome-wide association studies (GWAS), the mechanisms by which these variants operate remain largely unknown. Here we used 406 cancer ATAC-Seq samples across 23 cancer types to identify 7,262 germline allele-specific accessibility QTLs (as-aQTLs). Cancer as-aQTLs had stronger enrichment for cancer risk heritability (up to 145 fold) than any other functional annotation across seven cancer GWAS. Most cancer as-aQTLs directly altered transcription factor (TF) motifs and exhibited differential TF binding and gene expression in functional screens. To connect as-aQTLs to putative risk mechanisms, we introduced the regulome-wide associations study (RWAS). RWAS identified genetically associated accessible peaks at >70% of known breast and prostate loci and discovered new risk loci in all examined cancer types. Integrating as-aQTL discovery, motif analysis and RWAS identified candidate causal regulatory elements and their probable upstream regulators. Our work establishes cancer as-aQTLs and RWAS analysis as powerful tools to study the genetic architecture of cancer risk.

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: stratAS identifies as-aQTLs in cancer samples.
Fig. 2: Cancer as-aQTLs are enriched for cancer risk heritability and eQTLs.
Fig. 3: Cancer as-aQTLs are associated with differential TF binding and gene expression.
Fig. 4: RWAS links accessible chromatin regions to cancer risk.
Fig. 5: RWAS implicates hundreds of cancer risk mechanisms and outperforms TWAS.
Fig. 6: Breast cancer risk-associated RWAS peaks are linked to risk-associated TWAS genes.
Fig. 7: Allelic imbalance and RWAS can explain GWAS risk loci.

Similar content being viewed by others

Data availability

Full allelic imbalance results and all RWAS model weights are available at https://doi.org/10.5281/zenodo.6371439. ATAC-Seq data for all cancer samples are available at https://gdc.cancer.gov/about-data/publications/ATACseq-AWG. SNP-SELEX assay data is available at http://renlab.sdsc.edu/GVATdb/search.html. SuRE assay data is available at https://osf.io/6wev3/. The hg19 reference genome (human_g1k_v37) can be found at https://www.internationalgenome.org/category/grch37/. GTEx v.8 data can be found at https://www.gtexportal.org/home/datasets. Cancer eQTL data can be found at http://gong_lab.hzau.edu.cn/PancanQTL/. TWAS models can be found at http://gusevlab.org/projects/fusion/.

Code availability

Code to conduct allelic imbalance analyses, build RWAS models and conduct RWAS analyses is available at https://doi.org/10.5281/zenodo.6371678.

References

  1. Zhang, H. et al. Genome-wide association study identifies 32 novel breast cancer susceptibility loci from overall and subtype-specific analyses. Nat. Genet. 52, 572–581 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Michailidou, K. et al. Association analysis identifies 65 new breast cancer risk loci. Nature 551, 92–94 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Conti, D. V. et al. Trans-ancestry genome-wide association meta-analysis of prostate cancer identifies new susceptibility loci and informs genetic risk prediction. Nat. Genet. 53, 65–75 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. McKay, J. D. et al. Large-scale association analysis identifies new lung cancer susceptibility loci and heterogeneity in genetic susceptibility across histological subtypes. Nat. Genet. 49, 1126–1132 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Sud, A., Kinnersley, B. & Houlston, R. S. Genome-wide association studies of cancer: current insights and future perspectives. Nat. Rev. Cancer 17, 692–704 (2017).

    Article  CAS  PubMed  Google Scholar 

  6. Fachal, L. et al. Fine-mapping of 150 breast cancer risk regions identifies 191 likely target genes. Nat. Genet. 52, 56–73 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Gusev, A. et al. A transcriptome-wide association study of high-grade serous epithelial ovarian cancer identifies new susceptibility genes and splice variants. Nat. Genet. 51, 815–823 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Wu, L. et al. A transcriptome-wide association study of 229,000 women identifies new candidate susceptibility genes for breast cancer. Nat. Genet. 50, 968–978 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Mancuso, N. et al. Large-scale transcriptome-wide association study identifies new prostate cancer risk regions. Nat. Commun. 9, 4079 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Hormozdiari, F. et al. Leveraging molecular quantitative trait loci to understand the genetic architecture of diseases and complex traits. Nat. Genet. 50, 1041–1047 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. 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 

  12. Degner, J. F. et al. DNase I sensitivity QTLs are a major determinant of human expression variation. Nature 482, 390–394 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Gaffney, D. J. et al. Dissecting the regulatory architecture of gene expression QTLs. Genome Biol. 13, R7 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Battle, A. & Montgomery, S. B. Determining causality and consequence of expression quantitative trait loci. Hum. Genet. 133, 727–735 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. 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  PubMed  Google Scholar 

  16. Gong, J. et al. PancanQTL: systematic identification of cis-eQTLs and trans-eQTLs in 33 cancer types. Nucleic Acids Res. 46, D971–D976 (2018).

    Article  CAS  PubMed  Google Scholar 

  17. Geeleher, P. et al. Cancer expression quantitative trait loci (eQTLs) can be determined from heterogeneous tumor gene expression data by modeling variation in tumor purity. Genome Biol. 19, 130 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Li, Q. et al. Expression QTL-based analyses reveal candidate causal genes and loci across five tumor types. Hum. Mol. Genet. 23, 5294–5302 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. 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  PubMed  PubMed Central  Google Scholar 

  20. Mu, Z. et al. The impact of cell type and context-dependent regulatory variants on human immune traits. Genome Biol. 22, 122 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. 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 

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

    Article  CAS  PubMed  Google Scholar 

  23. Chun, S. et al. Limited statistical evidence for shared genetic effects of eQTLs and autoimmune-disease-associated loci in three major immune-cell types. Nat. Genet. 49, 600–605 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Waszak, S. M. et al. Population variation and genetic control of modular chromatin architecture in humans. Cell 162, 1039–1050 (2015).

    Article  CAS  PubMed  Google Scholar 

  25. Grubert, F. et al. Genetic control of chromatin states in humans involves local and distal chromosomal interactions. Cell 162, 1051–1065 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. McVicker, G. et al. Identification of genetic variants that affect histone modifications in human cells. Science 342, 747–749 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Gate, R. E. et al. Genetic determinants of co-accessible chromatin regions in activated T cells across humans. Nat. Genet. 50, 1140–1150 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Liang, D. et al. Cell-type-specific effects of genetic variation on chromatin accessibility during human neuronal differentiation. Nat. Neurosci. 24, 941–953 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Pickrell, J. K. et al. Understanding mechanisms underlying human gene expression variation with RNA sequencing. Nature 464, 768–772 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Yan, H., Yuan, W., Velculescu, V. E., Vogelstein, B. & Kinzler, K. W. Allelic variation in human gene expression. Science 297, 1143 (2002).

    Article  CAS  PubMed  Google Scholar 

  31. Battle, A. et al. Characterizing the genetic basis of transcriptome diversity through RNA-sequencing of 922 individuals. Genome Res. 24, 14–24 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Castel, S. E. et al. A vast resource of allelic expression data spanning human tissues. Genome Biol. 21, 234 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Wang, A. T. et al. Allele-specific QTL fine mapping with PLASMA. Am. J. Hum. Genet. 106, 170–187 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Liang, Y., Aguet, F., Barbeira, A. N., Ardlie, K. & Im, H. K. A scalable unified framework of total and allele-specific counts for cis-QTL, fine-mapping, and prediction. Nat. Commun. 12, 1424 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Gutierrez-Arcelus, M. et al. Allele-specific expression changes dynamically during T cell activation in HLA and other autoimmune loci. Nat. Genet. 52, 247–253 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Houlahan, K. E. et al. Genome-wide germline correlates of the epigenetic landscape of prostate cancer. Nat. Med. 25, 1615–1626 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Gusev, A. et al. Allelic imbalance reveals widespread germline-somatic regulatory differences and prioritizes risk loci in renal cell carcinoma. Preprint at bioRxiv https://doi.org/10.1101/631150 (2019).

  38. 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 

  39. Kumasaka, N., Knights, A. J. & Gaffney, D. J. Fine-mapping cellular QTLs with RASQUAL and ATAC-seq. Nat. Genet. 48, 206–213 (2016).

    Article  CAS  PubMed  Google Scholar 

  40. Corces, M. R. et al. The chromatin accessibility landscape of primary human cancers. Science 362, eaav1898 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Huyghe, J. R. et al. Discovery of common and rare genetic risk variants for colorectal cancer. Nat. Genet. 51, 76–87 (2019).

    Article  CAS  PubMed  Google Scholar 

  42. Schumacher, F. R. et al. Association analyses of more than 140,000 men identify 63 new prostate cancer susceptibility loci. Nat. Genet. 50, 928–936 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Scelo, G. et al. Genome-wide association study identifies multiple risk loci for renal cell carcinoma. Nat. Commun. 8, 15724 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Melin, B. S. et al. Genome-wide association study of glioma subtypes identifies specific differences in genetic susceptibility to glioblastoma and non-glioblastoma tumors. Nat. Genet. 49, 789–794 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Loh, P.-R., Kichaev, G., Gazal, S., Schoech, A. P. & Price, A. L. Mixed-model association for biobank-scale datasets. Nat. Genet. 50, 906–908 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Gazal, S., Marquez-Luna, C., Finucane, H. K. & Price, A. L. Reconciling S-LDSC and LDAK functional enrichment estimates. Nat. Genet. 51, 1202–1204 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  PubMed Central  Google Scholar 

  49. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Watt, A. C. et al. CDK4/6 inhibition reprograms the breast cancer enhancer landscape by stimulating AP-1 transcriptional activity. Nat. Cancer 2, 34–48 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Eferl, R. & Wagner, E. F. AP-1: a double-edged sword in tumorigenesis. Nat. Rev. Cancer 3, 859–868 (2003).

    Article  CAS  PubMed  Google Scholar 

  52. Verde, P., Casalino, L., Talotta, F., Yaniv, M. & Weitzman, J. B. Deciphering AP-1 function in tumorigenesis: fra-ternizing on target promoters. Cell Cycle 6, 2633–2639 (2007).

    Article  CAS  PubMed  Google Scholar 

  53. Kharman-Biz, A. et al. Expression of activator protein-1 (AP-1) family members in breast cancer. BMC Cancer 13, 441 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Tang, Y., Shu, G., Yuan, X., Jing, N. & Song, J. FOXA2 functions as a suppressor of tumor metastasis by inhibition of epithelial-to-mesenchymal transition in human lung cancers. Cell Res. 21, 316–326 (2011).

    Article  CAS  PubMed  Google Scholar 

  55. Parolia, A. et al. Distinct structural classes of activating FOXA1 alterations in advanced prostate cancer. Nature 571, 413–418 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Radhakrishnan, S. K. & Gartel, A. L. FOXM1: the Achilles’ heel of cancer? Nature reviews. Cancer vol. 8 c1; author reply c2 (2008).

  57. Chakrabarti, R. et al. Elf5 inhibits the epithelial-mesenchymal transition in mammary gland development and breast cancer metastasis by transcriptionally repressing Snail2. Nat. Cell Biol. 14, 1212–1222 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Peng, C. et al. Cyclin-dependent kinase 2 (CDK2) is a key mediator for EGF-induced cell transformation mediated through the ELK4/c-Fos signaling pathway. Oncogene 35, 1170–1179 (2016).

    Article  CAS  PubMed  Google Scholar 

  59. Cheng, M. et al. Transcription Factor ELF1 Activates MEIS1 Transcription and Then Regulates the GFI1/FBW7 Axis to Promote the Development of Glioma. Mol. Ther. Nucleic Acids 23, 418–430 (2021).

    Article  CAS  PubMed  Google Scholar 

  60. Jané-Valbuena, J. et al. An oncogenic role for ETV1 in melanoma. Cancer Res. 70, 2075–2084 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Pellecchia, A. et al. Overexpression of ETV4 is oncogenic in prostate cells through promotion of both cell proliferation and epithelial to mesenchymal transition. Oncogenesis 1, e20 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Miao, B. et al. The transcription factor FLI1 promotes cancer progression by affecting cell cycle regulation. Int. J. Cancer 147, 189–201 (2020).

    Article  CAS  PubMed  Google Scholar 

  63. Yan, J. Systematic analysis of binding of transcription factors to noncoding variants. Nature 591, 147–151 (2021).

    Article  CAS  PubMed  Google Scholar 

  64. van Arensbergen, J. et al. Genome-wide mapping of autonomous promoter activity in human cells. Nat. Biotechnol. 35, 145–153 (2017).

    Article  PubMed  CAS  Google Scholar 

  65. Gusev, A. et al. Integrative approaches for large-scale transcriptome-wide association studies. Nat. Genet. 48, 245–252 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Dadaev, T. et al. Fine-mapping of prostate cancer susceptibility loci in a large meta-analysis identifies candidate causal variants. Nat. Commun. 9, 2256 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Baca, S. C. et al. Genetic determinants of chromatin reveal prostate cancer risk mediated by context-dependent gene regulation. Preprint at bioRxiv https://doi.org/10.1101/2021.05.10.443466 (2021).

  68. Forbes, S. A. et al. COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res. 39, D945–D950 (2011).

    Article  CAS  PubMed  Google Scholar 

  69. Pasaniuc, B. et al. Enhanced statistical tests for GWAS in admixed populations: assessment using African Americans from CARe and a Breast Cancer Consortium. PLoS Genet. 7, e1001371 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Chimge, N.-O. & Frenkel, B. The RUNX family in breast cancer: relationships with estrogen signaling. Oncogene 32, 2121–2130 (2013).

    Article  CAS  PubMed  Google Scholar 

  71. Kalita, C. A. & Gusev, A. A novel method to identify cell-type specific regulatory variants and their role in cancer risk. bioRxiv https://doi.org/10.1101/2021.11.11.468278 (2021).

  72. Horn, S. et al. TERT promoter mutations in familial and sporadic melanoma. Science 339, 959–961 (2013).

    Article  CAS  PubMed  Google Scholar 

  73. Boix, C. A., James, B. T., Park, Y. P., Meuleman, W. & Kellis, M. Regulatory genomic circuitry of human disease loci by integrative epigenomics. Nature 590, 300–307 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Mancuso, N. et al. Integrating gene expression with summary association statistics to identify genes associated with 30 complex traits. Am. J. Hum. Genet. 100, 473–487 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. 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  PubMed  PubMed Central  Google Scholar 

  76. Strober, B. J. et al. Dynamic genetic regulation of gene expression during cellular differentiation. Science 364, 1287–1290 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. 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  PubMed  PubMed Central  CAS  Google Scholar 

  78. 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 

  79. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Broad Institute TCGA Genome Data Analysis Center. Analysis-ready standardized TCGA data from Broad GDAC Firehose 2016_01_28 run. (2016) https://doi.org/10.7908/C11G0KM9

  84. Friedman, J., Hastie, T. & Tibshirani, R. Regularization paths for generalized linear models via coordinate descent. J. Stat. Softw. 33, 1–22 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  85. Zhou, D. et al. A unified framework for joint-tissue transcriptome-wide association and Mendelian randomization analysis. Nat. Genet. 52, 1239–1246 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. 1000 Genomes Project Consortium. A global reference for human genetic variation. Nature 526, 68–74 (2015).

    Article  CAS  Google Scholar 

  87. Meyer, K. B. et al. Allele-specific up-regulation of FGFR2 increases susceptibility to breast cancer. PLoS Biol. 6, e108 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  88. Gusev, A. et al. Transcriptome-wide association study of schizophrenia and chromatin activity yields mechanistic disease insights. Nat. Genet. 50, 538–548 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Yang, J. et al. Conditional and joint multiple-SNP analysis of GWAS summary statistics identifies additional variants influencing complex traits. Nat. Genet. 44, 369–375 (2012). S1–S3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank M. Freedman, B. Pasaniuc and N. Mancuso for providing feedback on the manuscript. D.G. and A.G. were supported by R01 CA227237 and R01 CA244569. A.G. was also supported by R01 MH115676 and R01 CA259200. D.G. was also supported by the IBM Ph.D. Fellowship Award.

Author information

Authors and Affiliations

Authors

Contributions

A.G. conceived and supervised the project and developed RWAS. D.G. conducted all analyses. D.G. and A.G. wrote the manuscript.

Corresponding author

Correspondence to Alexander Gusev.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks Christopher Amos, Jason Stein and the other, anonymous, reviewer(s) 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 Effects of population structure, sample size, peak definitions and CNVs on imbalance analysis.

(a) Inferred genetic ancestry of ATAC-seq samples; each sample is a point color-coded by self-reported race (unfilled circles represent “not reported”) plotted along with the top two genetic Principal Components (PC) shown on the x and y axes. Allelic imbalance is not inflated by population structure due to “canceling out” trans/environmental effects by comparing functional activity at the two alleles within an individual, and only testing heterozygous carriers (thus not biased by population differences in allele frequency). (b) The number of discovered significant as-aQTLs (y-axis) grows linearly with the number of (randomly downsampled) cancer samples (x-axis). (c) The discovery of as-aQTLs does not strongly depend on peak calling with MACS. Many of the same regions are identified without any previous peak calling. (d) The density of as-aQTLs (y-axis) increases exponentially in high-CNV genomic regions (x-axis). The segment mean values are calculated from TCGA somatic copy number calls (segment mean =| log2(CNV/2)|). The chosen maximum threshold of 0.6 corresponds to the gain of one additional allele copy (0.58 ≈| log2(3/2) |).

Extended Data Fig. 2 Cancer as-aQTLs are strongly enriched for cancer risk heritability.

(a) Meta-analysis of 7 cancer types shows that as-aQTLs are more strongly enriched for cancer risk heritability (x-axis) than any other evaluated annotation (y-axis/bars); error bars correspond to the estimated standard error. Data are presented as inverse-variance weighted mean values + /- s.e. (b) as-aQTLs discovered in all 7 cancer types are also enriched for cancer risk variants when the enrichment is quantified using a simpler strategy based on top GWAS associations that does not account for background annotations (see Supplementary Note). Z-scores for the significance of enrichment are shown.

Extended Data Fig. 3 Heritability enrichment at cancer as-aQTLs is cancer type-specific.

(a-c) Cancer type-specific as-aQTLs do not exhibit heritability enrichment for noncancer traits. Data are presented as mean values + /- s.e. (d-e) The cancer risk heritability enrichment at pancancer as-aQTLs is lower than at matching cancer type-specific as-aQTLs. Data are presented as mean values + /- s.e.

Extended Data Fig. 4 Cancer as-aQTLs and d-as-aQTLs are strongly enriched for eQTLs.

(a) The enrichment of cancer-specific accessible peaks (by cancer type: y-axis) for intersecting GTEx tissue eQTLs (by tissue: x-axis) relative to random genomic sequences. Each cell reports the corresponding enrichment (ratio of eQTLs in peaks to eQTLs in random regions) and is shaded by this value. The pancancer peak set (top row) is expected to have a higher fraction of rare/private peaks, which likely explains the apparent lower eQTL enrichment. (b) The fraction of as-aQTLs (by cancer type: y-axis) that intersect one or more GTEx tissue eQTLs (by tissue: x-axis). Each cell reports the fraction and is shaded by this value. For many cancer-tissue pairs, >50% of as-aQTLs contain eQTLs. (c) The enrichment of cancer d-as-aQTLs for intersecting GTEx tissue eQTLs relative to all cancer type-specific peaks. Each cell reports the corresponding enrichment (ratio) and is shaded by this value. Non-significant enrichments (Z < 2, see Methods) are shown as NA. (d) The average Z-scores for the enrichment of cancer as-aQTLs for GTEx tissue eQTLs (averaged across all GTEx tissues, y-axis) as a function of the number of samples in the corresponding cancer type (x-axis). Each point is labeled by the corresponding cancer type. eQTL enrichment z-scores correlate with the number of samples per cancer type.

Extended Data Fig. 5 Distribution of allelic fractions at as-aQTLs with functional and disrupted TF motifs.

(a-l) Violin plots for selected TF motifs representing 12 motif families. The distribution of allelic fractions at as-aQTLs is shown for functional (red) and disrupted (blue) TF motifs.

Extended Data Fig. 6 Additional TF motif scores calculations at as-aQTLs and correlations between as-aQTL allelic fractions and TF binding / gene expression.

(a) Allelic fraction differences (left) and HOMER motif scores (right) from an additional 30 TF motifs for which the difference in motif scores between sequences with high and lower allelic fractions was most significant. Alleles with higher allelic fractions are shown in blue and alleles with lower allelic fractions in orange. Note the reversed directionality for the LRF motif, which is known to act as a repressor of transcription (Constantinou, C. et al. The multi-faceted functioning portrait of LRF/ZBTB7A. Hum. Genomics 13, 66 (2019)). Data are presented as mean values + /- s.d. (b) The correlation between significant SNP-SELEX SNP-TF pair PBS values (p < 0.01) and as-aQTL allelic fractions (y-axis) does not significantly change with varying allelic fraction thresholds |AF-0.5 | (x-axis). Data are presented as Pearson correlations + /- s.e. Number of pairs used for correlation analysis are shown above each point. (c) The correlation between significant SuRE SNP ΔExpressionALT-REF values (p < 0.00173121) and as-aQTL allelic fractions (y-axis) changes only slightly with varying allelic fraction thresholds |AF-0.5 | (x-axis). Data are presented as Pearson correlations + /- s.e. Number of pairs used for correlation analysis are shown above each point.

Extended Data Fig. 7 Additional RWAS analyses.

(a) Numbers of significant RWAS associations between peaks with variants and cancer risk discovered for 7 cancer types using 6 model types. (b) Numbers of significant RWAS associations between peaks without variants and cancer risk discovered for 7 cancer types using 6 model types. In total, 491036 peaks were analyzed of which 337793 (~69%) had a variant and 153243 (~31%) did not. (c) TWAS conducted using GTEx normal tissue expression data yields a comparable number of GWAS risk loci with TWAS genes. The lower number of prostate cancer TWAS genes is due to the smaller sample size of the normal prostate RNA-Seq dataset (132 GTEx samples vs. 468 TCGA samples). (d) RWAS still outperforms TWAS when only top1.total and top1.lasso model types are used (the same model types used for TWAS). (e) The correlations between heritable (cross-validation P of < 0.05) and TWAS-significant genes and RWAS-significant peaks at breast and prostate cancer risk loci are significantly stronger than the correlations between heritable (cross-validation p-values of < 0.05) but non-significant TWAS genes and RWAS peaks at GWAS risk loci. Horizontal lines inside the boxes indicate the medians. Box bounds show Q1 and Q3. Whiskers are minima (Q1 - 1.5x(Q3-Q1)) and maxima (Q3 + 1.5x(Q3-Q1)). (f) The correlation between heritable TWAS genes and RWAS peaks increases with decreasing distance. RWAS peak correlations with TWAS genes identified from expression in normal tissue are not significantly different from correlations with TWAS genes identified from the expression in cancer tissues. Matching TCGA breast normal and cancer samples were used for TWAS. Horizontal lines inside the boxes indicate the medians. Box bounds show Q1 and Q3. Whiskers are minima (Q1 - 1.5x(Q3-Q1)) and maxima (Q3 + 1.5x(Q3-Q1)).

Extended Data Fig. 8 Prostate cancer risk-associated RWAS peaks are linked to risk-associated TWAS genes.

(a) Correlations between TWAS gene and RWAS peak pairs at 33 prostate cancer GWAS risk loci. Nodes representing TWAS genes are shown in red with gene names shown in brackets. Nodes representing RWAS peaks are shown in black. The color of the edges represents the strength of the correlations between models (absolute Pearson correlation). (b) Median absolute Pearson correlations between significant TWAS gene and RWAS peak pairs at each GWAS prostate cancer risk locus. Horizontal lines inside the boxes indicate the medians. Box bounds show Q1 and Q3. Whiskers are minima (Q1 - 1.5x(Q3-Q1)) and maxima (Q3 + 1.5x(Q3-Q1)).

Extended Data Fig. 9 Prostate cancer risk-associated RWAS peaks are linked to risk-associated CWAS features.

(a) Overlap of significant RWAS and CWAS (H3K27ac) peaks with prostate cancer GWAS risk loci and significant RWAS and CWAS (AR) peaks with prostate cancer GWAS risk loci. (b) Correlations between prostate cancer risk-associated RWAS and CWAS peak pairs across all prostate cancer GWAS risk loci are similar to the RWAS-TWAS associations (Extended Data Fig. 8). Horizontal lines inside the boxes indicate the medians. Box bounds show Q1 and Q3. Whiskers are minima (Q1 - 1.5x(Q3-Q1)) and maxima (Q3 + 1.5x(Q3-Q1)). (c) Correlations between CWAS (H3K27ac) and RWAS peak pairs at 42 prostate cancer GWAS risk loci. Horizontal lines inside the boxes indicate the medians. Box bounds show Q1 and Q3. Whiskers are minima (Q1 - 1.5x(Q3-Q1)) and maxima (Q3 + 1.5x(Q3-Q1)). (d) Correlations between CWAS (AR) and RWAS peak pairs at 27 prostate cancer GWAS risk loci. Horizontal lines inside the boxes indicate the medians. Box bounds show Q1 and Q3. Whiskers are minima (Q1 - 1.5x(Q3-Q1)) and maxima (Q3 + 1.5x(Q3-Q1)).

Extended Data Fig. 10 RWAS associations can explain GWAS risk loci.

(a-f) Examples of RWAS-significant peaks near COSMIC genes that explain a large portion of the GWAS signal in conditional analyses. Each dot corresponds to a GWAS SNP with the significance of the association on the y-axis and physical position on the x-axis. The gray dots indicate marginal GWAS signals, and the blue dots show the same signals become less significant after conditioning on an as-aQTL/balanced peak identified in RWAS.

Supplementary information

Supplementary Information

Supplementary Notes and References

Reporting Summary

Supplementary Table 1

Supplementary Tables 1–8

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Grishin, D., Gusev, A. Allelic imbalance of chromatin accessibility in cancer identifies candidate causal risk variants and their mechanisms. Nat Genet 54, 837–849 (2022). https://doi.org/10.1038/s41588-022-01075-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-022-01075-2

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