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.

  • Resource
  • Published:

The reference epigenome and regulatory chromatin landscape of chronic lymphocytic leukemia

Abstract

Chronic lymphocytic leukemia (CLL) is a frequent hematological neoplasm in which underlying epigenetic alterations are only partially understood. Here, we analyze the reference epigenome of seven primary CLLs and the regulatory chromatin landscape of 107 primary cases in the context of normal B cell differentiation. We identify that the CLL chromatin landscape is largely influenced by distinct dynamics during normal B cell maturation. Beyond this, we define extensive catalogues of regulatory elements de novo reprogrammed in CLL as a whole and in its major clinico-biological subtypes classified by IGHV somatic hypermutation levels. We uncover that IGHV-unmutated CLLs harbor more active and open chromatin than IGHV-mutated cases. Furthermore, we show that de novo active regions in CLL are enriched for NFAT, FOX and TCF/LEF transcription factor family binding sites. Although most genetic alterations are not associated with consistent epigenetic profiles, CLLs with MYD88 mutations and trisomy 12 show distinct chromatin configurations. Furthermore, we observe that non-coding mutations in IGHV-mutated CLLs are enriched in H3K27ac-associated regulatory elements outside accessible chromatin. Overall, this study provides an integrative portrait of the CLL epigenome, identifies extensive networks of altered regulatory elements and sheds light on the relationship between the genetic and epigenetic architecture of the 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: CLL reference epigenomes.
Fig. 2: Chromatin states and its transitions in CLL.
Fig. 3: CLL specific regulatory landscape.
Fig. 4: De novo chromatin activity and accessibility changes in an extended CLL cohort.
Fig. 5: B cell related chromatin activity and accessibility signatures in the extended CLL cohort.
Fig. 6: Somatic genetic alterations in relation to chromatin activity and accessibility.

Similar content being viewed by others

References

  1. Baylin, S. B. & Jones, P. A. A decade of exploring the cancer epigenome—Biological and translational implications. Nat. Rev. Cancer 11, 726–734 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Rivera, C. M. & Ren, B. Mapping human epigenomes. Cell 155, 39–55 (2013).

    Article  PubMed  CAS  Google Scholar 

  3. Akhtar-Zaidi, B. et al. Epigenomic enhancer profiling defines a signature of colon cancer. Science 336, 736–739 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Fiziev, P. et al. Systematic epigenomic analysis reveals chromatin states associated with melanoma progression. Cell Rep. 19, 875–889 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Lin, C. Y. et al. Active medulloblastoma enhancers reveal subgroup-specific cellular origins. Nature 530, 57–62 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Muratani, M. et al. Nanoscale chromatin profiling of gastric adenocarcinoma reveals cancer-associated cryptic promoters and somatically acquired regulatory elements. Nat. Commun. 5, 4361 (2014).

    Article  PubMed  CAS  Google Scholar 

  7. Queiros, A. C. et al. Decoding the DNA methylome of mantle cell lymphoma in the light of the entire B cell lineage. Cancer Cell 30, 806–821 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Rendeiro, A. F. et al. Chromatin accessibility maps of chronic lymphocytic leukaemia identify subtype-specific epigenome signatures and transcription regulatory networks. Nat. Commun. 7, 11938 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Chun, H. J. et al. Genome-wide profiles of extra-cranial malignant rhabdoid tumors reveal heterogeneity and dysregulated developmental pathways. Cancer Cell 29, 394–406 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Khurana, E. et al. Role of non-coding sequence variants in cancer. Nat. Rev. Genet. 17, 93–108 (2016).

    Article  PubMed  CAS  Google Scholar 

  11. Shen, H. & Laird, P. W. Interplay between the cancer genome and epigenome. Cell 153, 38–55 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Fabbri, G. & Dalla-Favera, R. The molecular pathogenesis of chronic lymphocytic leukaemia. Nat. Rev. Cancer 16, 145–162 (2016).

    Article  PubMed  CAS  Google Scholar 

  13. Kipps, T. J. et al. Chronic lymphocytic leukaemia. Nat. Rev. Dis. Primers 3, 16096 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Damle, R. N. et al. Ig V gene mutation status and CD38 expression as novel prognostic indicators in chronic lymphocytic leukemia. Blood 94, 1840–1847 (1999).

    PubMed  CAS  Google Scholar 

  15. Hamblin, T. J., Davis, Z., Gardiner, A., Oscier, D. G. & Stevenson, F. K. Unmutated Ig V(H) genes are associated with a more aggressive form of chronic lymphocytic leukemia. Blood 94, 1848–1854 (1999).

    PubMed  CAS  Google Scholar 

  16. Kulis, M. et al. Epigenomic analysis detects widespread gene-body DNA hypomethylation in chronic lymphocytic leukemia. Nat. Genet. 44, 1236–1242 (2012).

    Article  PubMed  CAS  Google Scholar 

  17. Landau, D. A. et al. Locally disordered methylation forms the basis of intratumor methylome variation in chronic lymphocytic leukemia. Cancer Cell 26, 813–825 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Landau, D. A. et al. Mutations driving CLL and their evolution in progression and relapse. Nature 526, 525–530 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Oakes, C. C. et al. DNA methylation dynamics during B cell maturation underlie a continuum of disease phenotypes in chronic lymphocytic leukemia. Nat. Genet. 48, 253–264 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Puente, X. S. et al. Non-coding recurrent mutations in chronic lymphocytic leukaemia. Nature 526, 519–524 (2015).

    Article  PubMed  CAS  Google Scholar 

  21. Cahill, N. et al. 450K-array analysis of chronic lymphocytic leukemia cells reveals global DNA methylation to be relatively stable over time and similar in resting and proliferative compartments. Leukemia 27, 150–158 (2013).

    Article  PubMed  CAS  Google Scholar 

  22. Ferreira, P. G. et al. Transcriptome characterization by RNA sequencing identifies a major molecular and clinical subdivision in chronic lymphocytic leukemia. Genome Res. 24, 212–226 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. International Cancer Genome, C. et al. International network of cancer genome projects. Nature 464, 993–998 (2010).

    Article  CAS  Google Scholar 

  24. Kulis, M. et al. Whole-genome fingerprint of the DNA methylome during human B cell differentiation. Nat. Genet. 47, 746–756 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Ernst, J. et al. Mapping and analysis of chromatin state dynamics in nine human cell types. Nature 473, 43–49 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Ernst, J. & Kellis, M. ChromHMM: Automating chromatin-state discovery and characterization. Nat. Methods 9, 215–216 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Hnisz, D. et al. Super-enhancers in the control of cell identity and disease. Cell 155, 934–947 (2013).

    Article  PubMed  CAS  Google Scholar 

  28. Seifert, M. et al. Cellular origin and pathophysiology of chronic lymphocytic leukemia. J. Exp. Med. 209, 2183–2198 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Jin, F. et al. A high-resolution map of the three-dimensional chromatin interactome in human cells. Nature 503, 290–294 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Javierre, B. M. et al. Lineage-specific genome architecture links enhancers and non-coding disease variants to target gene promoters. Cell 167, 1369–1384 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. McCarthy, B. A. et al. A seven-gene expression panel distinguishing clonal expansions of pre-leukemic and chronic lymphocytic leukemia B cells from normal B lymphocytes. Immunol. Res. 63, 90–100 (2015).

    Article  PubMed  CAS  Google Scholar 

  33. Navarro, A., et al. Improved classification of leukemic B-cell lymphoproliferative disorders using a transcriptional and genetic classifier. Haematologica (2017).

  34. Gutierrez, A. Jr. et al. LEF-1 is a prosurvival factor in chronic lymphocytic leukemia and is expressed in the preleukemic state of monoclonal B-cell lymphocytosis. Blood 116, 2975–2983 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Ceribelli, M. et al. A druggable TCF4- and BRD4-dependent transcriptional network sustains malignancy in blastic plasmacytoid dendritic cell neoplasm. Cancer Cell 30, 764–778 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Queiros, A. C. et al. A B-cell epigenetic signature defines three biologic subgroups of chronic lymphocytic leukemia with clinical impact. Leukemia 29, 598–605 (2015).

    Article  PubMed  CAS  Google Scholar 

  37. Khandanpour, C. et al. Growth factor independence 1 antagonizes a p53-induced DNA damage response pathway in lymphoblastic leukemia. Cancer Cell 23, 200–214 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Moroy, T. & Khandanpour, C. Growth factor independence 1 (Gfi1) as a regulator of lymphocyte development and activation. Semin. Immunol. 23, 368–378 (2011).

    Article  PubMed  CAS  Google Scholar 

  39. Murphy, E. J. et al. Leukemia-cell proliferation and disease progression in patients with early stage chronic lymphocytic leukemia. Leukemia 31, 1348–1354 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Bachmaier, K. et al. Negative regulation of lymphocyte activation and autoimmunity by the molecular adaptor Cbl-b. Nature 403, 211–216 (2000).

    Article  PubMed  CAS  Google Scholar 

  41. Nihira, K. et al. Pim-1 controls NF-kappaB signalling by stabilizing RelA/p65. Cell Death Differ. 17, 689–698 (2010).

    Article  PubMed  CAS  Google Scholar 

  42. Kasof, G. M. et al. Tumor necrosis factor-alpha induces the expression of DR6, a member of the TNF receptor family, through activation of NF-kappaB. Oncogene 20, 7965–7975 (2001).

    Article  PubMed  CAS  Google Scholar 

  43. Xia, X. Z. et al. TACI is a TRAF-interacting receptor for TALL-1, a tumor necrosis factor family member involved in B cell regulation. J. Exp. Med. 192, 137–143 (2000).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Minici, C. et al. Distinct homotypic B-cell receptor interactions shape the outcome of chronic lymphocytic leukaemia. Nat. Commun. 8, 15746 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Schuster-Bockler, B. & Lehner, B. Chromatin organization is a major influence on regional mutation rates in human cancer cells. Nature 488, 504–507 (2012).

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Pedersen, B. S. & Quinlan, A. R. Who’s who? Detecting and resolving sample anomalies in human DNA sequencing studies with peddy. Am. J. Hum. Genet. 100, 406–413 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Wolf, C. et al. NFATC1 activation by DNA hypomethylation in chronic lymphocytic leukemia correlates with clinical staging and can be inhibited by ibrutinib. Int. J. Cancer 142, 322–333 (2018).

    Article  PubMed  CAS  Google Scholar 

  49. Wu, W. et al. High LEF1 expression predicts adverse prognosis in chronic lymphocytic leukemia and may be targeted by ethacrynic acid. Oncotarget 7, 21631–21643 (2016).

    PubMed  PubMed Central  Google Scholar 

  50. Jones, P. A., Issa, J. P. & Baylin, S. Targeting the cancer epigenome for therapy. Nat. Rev. Genet. 17, 630–641 (2016).

    Article  PubMed  CAS  Google Scholar 

  51. Loven, J. et al. Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153, 320–334 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Riches, J. C. et al. Trisomy 12 chronic lymphocytic leukemia cells exhibit upregulation of integrin signaling that is modulated by NOTCH1 mutations. Blood 123, 4101–4110 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Martinez-Trillos, A. et al. Clinical impact of MYD88 mutations in chronic lymphocytic leukemia. Blood 127, 1611–1613 (2016).

    Article  PubMed  CAS  Google Scholar 

  54. Burns, A., et al. Whole-genome sequencing of chronic lymphocytic leukaemia reveals distinct differences in the mutational landscape between IgHVmut and IgHVunmut subgroups. Leukemia (2017).

  55. Qian, J. et al. B cell super-enhancers and regulatory clusters recruit AID tumorigenic activity. Cell 159, 1524–1537 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Ecker, S. et al. Genome-wide analysis of differential transcriptional and epigenetic variability across human immune cell types. Genome Biol. 18, 18 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. van de Werken, H. J. et al. Robust 4C-seq data analysis to screen for regulatory DNA interactions. Nat. Methods 9, 969–972 (2012).

    Article  PubMed  CAS  Google Scholar 

  58. Simonis, M., Kooren, J. & de Laat, W. An evaluation of 3C-based methods to capture DNA interactions. Nat. Methods 4, 895–901 (2007).

    Article  PubMed  CAS  Google Scholar 

  59. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Leek, J.T. svaseq: Removing batch effects and other unwanted noise from sequencing data. Nucleic Acids Res. 42, e161 (2014).

    Article  PubMed Central  CAS  Google Scholar 

  63. Cairns, J. et al. CHiCAGO: Robust detection of DNA looping interactions in Capture Hi-C data. Genome Biol. 17, 127 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Serra, F., Baù, D., Filion, G. & Marti-Renom, M.A. Structural features of the fly chromatin colors revealed by automatic three-dimensional modeling. bioRxiv, https://doi.org/10.1101/036764 (2016).

  65. Ay, F. et al. Identifying multi-locus chromatin contacts in human cells using tethered multiple 3C. BMC Genomics 16, 121 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Thongjuea, S., Stadhouders, R., Grosveld, F. G. & Soler, E. & Lenhard, B. r3Cseq: An R/Bioconductor package for the discovery of long-range genomic interactions from chromosome conformation capture and next-generation sequencing data. Nucleic Acids Res. 41, e132 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Van der Auwera, G. A. et al. From FastQ data to high confidence variant calls: The Genome Analysis Toolkit best practices pipeline. Curr. Protoc. Bioinformatics 43, 11.10.1–11.10.33 (2013).

    Google Scholar 

  69. Merkel, A., et al. GEMBS—High through-put processing for DNA methylation data from whole genome bisulfite sequencing (WGBS). bioRxiv (2017).

  70. Garrison, E. & Marth, G. Haplotype-based variant detection from short-read sequencing. arXiv https://arxiv.org/abs/1207.3907 (2012).

  71. Juhling, F. et al. metilene: Fast and sensitive calling of differentially methylated regions from bisulfite sequencing data. Genome Res. 26, 256–262 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. de Wit, E. et al. The pluripotent genome in three dimensions is shaped around pluripotency factors. Nature 501, 227–231 (2013).

    Article  PubMed  CAS  Google Scholar 

  73. Falcon, S. & Gentleman, R. Using GOstats to test gene lists for GO term association. Bioinformatics 23, 257–258 (2007).

    Article  PubMed  CAS  Google Scholar 

  74. McLeay, R. C. & Bailey, T. L. Motif Enrichment Analysis: A unified framework and an evaluation on ChIP data. BMC Bioinformatics 11, 165 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Mathelier, A. et al. JASPAR 2016: A major expansion and update of the open-access database of transcription factor binding profiles. Nucleic Acids Res. 44, D110–115 (2016).

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

This work was funded by the European Union’s Seventh Framework Programme through the Blueprint Consortium (grant agreement 282510), the International Cancer Genome Consortium (Chronic Lymphocytic Leukemia Genome consortium to E.C. and C.L.-O.), the European Hematology Association (Non-Clinical Advanced Research Fellowship to J.I.M.-S.), the World Wide Cancer Research Foundation Grant No. 16-1285 (to J.I.M.-S.), the Spanish Ministerio de Economía y Competitividad (MINECO) Grant No. SAF2015-64885-R (to E.C.) and Grant No. PMP15/00007, part of Plan Nacional de I+D+I and co-financed by the ISCIII-Sub-Directorate General for Evaluation and the European Regional Development Fund (FEDER–“Una manera de hacer Europa”) (to E.C.), the Generalitat de Catalunya Suport Grups de Recerca AGAUR 2014-SGR-795 (to E.C.), the CERCA Programme/Generalitat de Catalunya and CIBERONC. R.B. was supported by fellowships from the EU (Marie Skłodowska-Curie Inter European Fellowship) and the Lady TATA Memorial Trust (International Award), N.R. by the Acció instrumental d’incorporació de científics i tecnòlegs PERIS 2016 from the Generalitat de Catalunya and M.Ku. by an AOI grant of the Spanish Association Against Cancer. E.C. is an Academia Researcher of the “Institució Catalana de Recerca i Estudis Avançats” (ICREA) of the Generalitat de Catalunya. This work was partially developed at the Centro Esther Koplowitz (CEK, Barcelona, Spain). We are indebted to the HCB-IDIBAPS Biobank-Tumor Bank and Hematopathology Collection for sample procurement.

Author information

Authors and Affiliations

Authors

Contributions

The Chronic Lymphocytic Leukemia Genome consortium and the BLUEPRINT consortium contributed to this study respectively as part of the International Cancer Genome Consortium and the International Human Epigenome Consortium. Investigator contributions were as follows: T.B., J.D., A.L.-G., D.M.-G., S.B., M.P., M.A., M.Ku., N.V.-D., X.A. and F.P. contributed to sample collection (CLL and normal B cells) as well as to their biological and clinical annotation. M.P., N.V.-D., M.G. and I.G. contributed to WGS data generation. N.R., N.V.-D., J.H.A.M., H.G.S., J.I.M.-S., M.G., I.G. and M.-L.Y. contributed to histone mark, ATAC-seq, methylome and transcriptome data generation. R.V.-B., J.B., M.G., I.G., J.I.M.-S., B.M.J., P.Fr., N.V.-D., A.E., A.C.Q. and R.B. contributed to in situ HiC, promoter capture HiC and 4C-seq data generation. X.S.P. and C.L.-O. contributed to WGS data analysis. R.B., V.C., J.H.A.M., M.D.-F., M.Ku., G.Cl., G.Ca., A.M., S.H., A.V., S.U., E.P., R.G., R.R., M.P., D.T., A.D., E.L., M.Ko., M.R., L.C., P.Fl. and J.I.M.-S. contributed to histone mark, ATAC-seq, methylome and transcriptome data analysis. R.V.-B., F.S., M.A.M.-R., S.W.W., B.M.J., P.Fr. and R.B. contributed to in situ HiC, promoter capture HiC and 4C-seq data analysis. C.L.-O., E.C., H.G.S. and R.S. participated in the study design and data interpretation together with R.B. and J.I.M.-S. R.B. and J.I.M.-S. directed the research and wrote the manuscript.

Corresponding author

Correspondence to Jose I. Martin-Subero.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Supplementary information

Supplementary Figures

Supplementary Figures 1-19

Reporting Summary

Supplementary Table 1

Patient characteristics

Supplementary Table 2

K-means clusters

Supplementary Table 3

Differential DNA methylation in CLL

Supplementary Table 4

Genes related to K-means clusters histone marks

Supplementary Table 5

Gene ontology terms

Supplementary Table 6

Chromatin states related to K-means clusters histone marks

Supplementary Table 7

De novo active and inactive regulatory elements in CLL

Supplementary Table 8

Enriched transcription factor motifs

Supplementary Table 9

Differentially active/accessible regions U-CLL and M-CLL

Supplementary Table 10

Overlap cell of origin signature DNA methylation and ATACseq

Supplementary Table 11

Patient groups somatic mutations and structural variants

Supplementary Table 12

Differential regions and target genes related to somatic genetic

Supplementary Table 13

Regions considered IG or SHM target loci in this study

Supplementary Table 14

Data quality measures

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Beekman, R., Chapaprieta, V., Russiñol, N. et al. The reference epigenome and regulatory chromatin landscape of chronic lymphocytic leukemia. Nat Med 24, 868–880 (2018). https://doi.org/10.1038/s41591-018-0028-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-018-0028-4

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