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.

  • Letter
  • Published:

DNMT3A mutations promote anthracycline resistance in acute myeloid leukemia via impaired nucleosome remodeling

Abstract

Although the majority of patients with acute myeloid leukemia (AML) initially respond to chemotherapy, many of them subsequently relapse, and the mechanistic basis for AML persistence following chemotherapy has not been determined. Recurrent somatic mutations in DNA methyltransferase 3A (DNMT3A), most frequently at arginine 882 (DNMT3AR882), have been observed in AML1,2,3 and in individuals with clonal hematopoiesis in the absence of leukemic transformation4,5. Patients with DNMT3AR882 AML have an inferior outcome when treated with standard-dose daunorubicin-based induction chemotherapy6,7, suggesting that DNMT3AR882 cells persist and drive relapse8. We found that Dnmt3a mutations induced hematopoietic stem cell expansion, cooperated with mutations in the FMS-like tyrosine kinase 3 gene (Flt3ITD) and the nucleophosmin gene (Npm1c) to induce AML in vivo, and promoted resistance to anthracycline chemotherapy. In patients with AML, the presence of DNMT3AR882 mutations predicts minimal residual disease, underscoring their role in AML chemoresistance. DNMT3AR882 cells showed impaired nucleosome eviction and chromatin remodeling in response to anthracycline treatment, which resulted from attenuated recruitment of histone chaperone SPT-16 following anthracycline exposure. This defect led to an inability to sense and repair DNA torsional stress, which resulted in increased mutagenesis. Our findings identify a crucial role for DNMT3AR882 mutations in driving AML chemoresistance and highlight the importance of chromatin remodeling in response to cytotoxic chemotherapy.

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

Figure 1: Dnmt3aR878H mutation augments HSC function and cooperates with co-occurring AML disease alleles in vivo.
Figure 2: Expression of mutant DNMT3A leads to anthracycline resistance.
Figure 3: Cells with mutant DNMT3A have a DNA damage signaling defect in response to anthracyclines.
Figure 4: Expression of mutant DNMT3A impairs chromatin remodeling in response to DNA distortion.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Cancer Genome Atlas Research Network. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N. Engl. J. Med. 368, 2059–2074 (2013).

  2. Ley, T.J. et al. DNMT3A mutations in acute myeloid leukemia. N. Engl. J. Med. 363, 2424–2433 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Yan, X.J. et al. Exome sequencing identifies somatic mutations of DNA methyltransferase gene DNMT3A in acute monocytic leukemia. Nat. Genet. 43, 309–315 (2011).

    Article  CAS  PubMed  Google Scholar 

  4. Genovese, G. et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N. Engl. J. Med. 371, 2477–2487 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Jaiswal, S. et al. Age-related clonal hematopoiesis associated with adverse outcomes. N. Engl. J. Med. 371, 2488–2498 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Patel, J.P. et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N. Engl. J. Med. 366, 1079–1089 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Sehgal, A.R. et al. DNMT3A mutational status affects the results of dose-escalated induction therapy in acute myelogenous leukemia. Clin. Cancer Res. 21, 1614–1620 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Shlush, L.I. et al. Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature 506, 328–333 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Leiserson, M.D. et al. Pan-cancer network analysis identifies combinations of rare somatic mutations across pathways and protein complexes. Nat. Genet. 47, 106–114 (2015).

    Article  CAS  PubMed  Google Scholar 

  10. Welch, J.S. et al. The origin and evolution of mutations in acute myeloid leukemia. Cell 150, 264–278 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Busque, L. et al. Recurrent somatic TET2 mutations in normal elderly individuals with clonal hematopoiesis. Nat. Genet. 44, 1179–1181 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Jan, M. et al. Clonal evolution of preleukemic hematopoietic stem cells precedes human acute myeloid leukemia. Sci. Transl. Med. 4, 149ra118 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Challen, G.A. et al. Dnmt3a is essential for hematopoietic stem cell differentiation. Nat. Genet. 44, 23–31 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Moran-Crusio, K. et al. Tet2 loss leads to increased hematopoietic stem cell self-renewal and myeloid transformation. Cancer Cell 20, 11–24 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Quivoron, C. et al. TET2 inactivation results in pleiotropic hematopoietic abnormalities in mouse and is a recurrent event during human lymphomagenesis. Cancer Cell 20, 25–38 (2011).

    Article  CAS  PubMed  Google Scholar 

  16. Li, Z. et al. Deletion of Tet2 in mice leads to dysregulated hematopoietic stem cells and subsequent development of myeloid malignancies. Blood 118, 4509–4518 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Yang, L., Rau, R. & Goodell, M.A. DNMT3A in haematological malignancies. Nat. Rev. Cancer 15, 152–165 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Holz-Schietinger, C., Matje, D.M. & Reich, N.O. Mutations in DNA methyltransferase (DNMT3A) observed in acute myeloid leukemia patients disrupt processive methylation. J. Biol. Chem. 287, 30941–30951 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Russler-Germain, D.A. et al. The R882H DNMT3A mutation associated with AML dominantly inhibits wild-type DNMT3A by blocking its ability to form active tetramers. Cancer Cell 25, 442–454 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kim, S.J. et al. A DNMT3A mutation common in AML exhibits dominant-negative effects in murine ES cells. Blood 122, 4086–4089 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Papaemmanuil, E. et al. Genomic classification and prognosis in acute myeloid leukemia. N. Engl. J. Med. 374, 2209–2221 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Gaidzik, V.I. et al. Clinical impact of DNMT3A mutations in younger adult patients with acute myeloid leukemia: results of the AML Study Group (AMLSG). Blood 121, 4769–4777 (2013).

    Article  CAS  PubMed  Google Scholar 

  23. Ostronoff, F. et al. Mutations in the DNMT3A exon 23 independently predict poor outcome in older patients with acute myeloid leukemia: a SWOG report. Leukemia 27, 238–241 (2013).

    Article  CAS  PubMed  Google Scholar 

  24. Ribeiro, A.F. et al. Mutant DNMT3A: a marker of poor prognosis in acute myeloid leukemia. Blood 119, 5824–5831 (2012).

    Article  CAS  PubMed  Google Scholar 

  25. Tie, R. et al. Association between DNMT3A mutations and prognosis of adults with de novo acute myeloid leukemia: a systematic review and meta-analysis. PLoS One 9, e93353 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Chen, X. et al. Relation of clinical response and minimal residual disease and their prognostic impact on outcome in acute myeloid leukemia. J. Clin. Oncol. 33, 1258–1264 (2015).

    Article  PubMed  Google Scholar 

  27. Ganzel, C. et al. Minimal residual disease assessment by flow cytometry in AML is an independent prognostic factor even after adjusting for cytogenetic/molecular abnormalities. Blood 124, 1016 (2014).

    Article  Google Scholar 

  28. Ivey, A. et al. Assessment of minimal residual disease in standard-risk AML. N. Engl. J. Med. 374, 422–433 (2016).

    Article  CAS  PubMed  Google Scholar 

  29. Othus, M. et al. Effect of measurable ('minimal') residual disease (MRD) information on prediction of relapse and survival in adult acute myeloid leukemia. Leukemia 30, 2080–2083 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Walter, R.B. et al. Comparison of minimal residual disease as outcome predictor for AML patients in first complete remission undergoing myeloablative or nonmyeloablative allogeneic hematopoietic cell transplantation. Leukemia 29, 137–144 (2015).

    Article  CAS  PubMed  Google Scholar 

  31. Shih, A.H. et al. Mutational cooperativity linked to combinatorial epigenetic gain of function in acute myeloid leukemia. Cancer Cell 27, 502–515 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Akalin, A. et al. Base-pair resolution DNA methylation sequencing reveals profoundly divergent epigenetic landscapes in acute myeloid leukemia. PLoS Genet. 8, e1002781 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Garrett-Bakelman, F.E. et al. Enhanced reduced representation bisulfite sequencing for assessment of DNA methylation at base pair resolution. J. Vis. Exp. http://dx.doi.org/10.3791/52246, e52246 (2015).

  34. Yang, F., Teves, S.S., Kemp, C.J. & Henikoff, S. Doxorubicin, DNA torsion, and chromatin dynamics. Biochim. Biophys. Acta 1845, 84–89 (2014).

    CAS  PubMed  Google Scholar 

  35. Stracker, T.H., Usui, T. & Petrini, J.H. Taking the time to make important decisions: the checkpoint effector kinases Chk1 and Chk2 and the DNA damage response. DNA Repair (Amst.) 8, 1047–1054 (2009).

    Article  CAS  Google Scholar 

  36. Collins, A.R. et al. The comet assay: topical issues. Mutagenesis 23, 143–151 (2008).

    Article  CAS  PubMed  Google Scholar 

  37. Pang, B. et al. Drug-induced histone eviction from open chromatin contributes to the chemotherapeutic effects of doxorubicin. Nat. Commun. 4, 1908 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Dinant, C. et al. Enhanced chromatin dynamics by FACT promotes transcriptional restart after UV-induced DNA damage. Mol. Cell 51, 469–479 (2013).

    Article  CAS  PubMed  Google Scholar 

  39. Ransom, M., Dennehey, B.K. & Tyler, J.K. Chaperoning histones during DNA replication and repair. Cell 140, 183–195 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Belotserkovskaya, R. et al. FACT facilitates transcription-dependent nucleosome alteration. Science 301, 1090–1093 (2003).

    Article  CAS  PubMed  Google Scholar 

  41. Mayle, A., Luo, M., Jeong, M. & Goodell, M.A. Flow cytometry analysis of murine hematopoietic stem cells. Cytometry A 83, 27–37 (2013).

    Article  CAS  PubMed  Google Scholar 

  42. Akashi, K., Traver, D., Miyamoto, T. & Weissman, I.L. A clonogenic common myeloid progenitor that gives rise to all myeloid lineages. Nature 404, 193–197 (2000).

    Article  CAS  PubMed  Google Scholar 

  43. Chen, K. et al. Resolving the distinct stages in erythroid differentiation based on dynamic changes in membrane protein expression during erythropoiesis. Proc. Natl. Acad. Sci. USA 106, 17413–17418 (2009).

    Article  PubMed  Google Scholar 

  44. Socolovsky, M. et al. Ineffective erythropoiesis in Stat5a−/−5b−/− mice due to decreased survival of early erythroblasts. Blood 98, 3261–3273 (2001).

    Article  CAS  PubMed  Google Scholar 

  45. Paietta, E. Should minimal residual disease guide therapy in AML? Best Pract. Res. Clin. Haematol. 28, 98–105 006 (2015).

    Article  PubMed  Google Scholar 

  46. Paietta, E. Minimal residual disease in acute myeloid leukemia: coming of age. Hematology (Am Soc Hematol Educ Program) 2012, 35–42 (2012).

    Article  Google Scholar 

  47. Cheng, D.T. et al. Memorial Sloan Kettering-integrated mutation profiling of actionable cancer targets (MSK-IMPACT): A hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology. J. Mol. Diagn. 17, 251–264 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Nguyen, S., Meletis, K., Fu, D., Jhaveri, S. & Jaenisch, R. Ablation of de novo DNA methyltransferase Dnmt3a in the nervous system leads to neuromuscular defects and shortened lifespan. Dev. Dyn. 236, 1663–1676 (2007).

    Article  CAS  PubMed  Google Scholar 

  49. Havre, P.A., Yuan, J., Hedrick, L., Cho, K.R. & Glazer, P.M. p53 inactivation by HPV16 E6 results in increased mutagenesis in human cells. Cancer Res. 55, 4420–4424 (1995).

    CAS  PubMed  Google Scholar 

  50. Whitfield, M.L. et al. Identification of genes periodically expressed in the human cell cycle and their expression in tumors. Mol. Biol. Cell 13, 1977–2000 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Wysocka, J. Identifying novel proteins recognizing histone modifications using peptide pull-down assay. Methods 40, 339–343 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Westfall, P.H. & Young, S.S. P-value adjustments for multiple tests in multivariate binomial models. J. Am. Stat. Assoc. 84, 780–786 (1989).

    Google Scholar 

Download references

Acknowledgements

The authors are grateful to A. Ringler, C. Sheridan, J. Gandara and Y. Neelamraju for technical support; to Weill Cornell Medicine Epigenomics Core for sequencing services; to H. Erdjument-Bromage for assistance with proteomics; to S. Zha (Columbia University Medical Center); and to A. Ciccia (Columbia University Medical Center) for critical reading of the manuscript and helpful suggestions. cDNA encoding human wild-type DNMT3A was a kind gift from F. Fuks (Free University of Brussels). This work was supported by NCI K99 grant CA178191 and Lauri Strauss Leukemia Foundation award to O.A.G.; by NCI K08 grant CA169055 and an American Society of Hematology (ASHAMFDP-20121) under the ASH-AMFDP partnership with The Robert Wood Johnson Foundation to F.E.G.-B.; by a Hyundai Hope On Wheels award to B.S.; by a Gabrielle's Angel Fund grant to R.L.L. and A.M.M.; by NCI grant CA172636 to R.L.L. and A.M.M.; by the Samuel Waxman Cancer Research Center; by a Stand Up To Cancer Convergence Award to R.L.L; and the NCI U10 grant CA180827 to E.M.P., R.L.L. and A.M.M. A.S.M. is supported by NIH grants T32GM007739 and F30CA18349. A.M.M. is a Burroughs Wellcome Clinical Translational Scholar, and is supported by the Sackler Center for Biomedical and Physical Sciences. R.L.L. is a LLS Scholar. P.B.S. is supported by the Austrian Research Foundation (#P27132) and the Oesterreichische Nationalbank (OeNB) Anniversary Fund (#15936). MSKCC cores used in these studies are supported by the P30 Core Grant CA008748. A part of this study was coordinated by the ECOG-ACRIN Cancer Research Group (R.L. Comis and M.D. Schnall) and supported in part by Public Health Service Grants CA180820, CA180794, CA180791, CA189859, CA180827 and from the National Cancer Institute, National Institutes of Health and the Department of Health and Human Services. Its content is solely the responsibility of the authors and does not necessarily represent the official views of the National Cancer Institute.

Author information

Authors and Affiliations

Authors

Contributions

O.A.G. and K.S. performed most of the experiments. L.B., M.D.K., D.T., E.L., A.R.W., A.R.G., E.P., F.P. and A.S.M. assisted with many of the experiments. C.Z., J.M.R., M.S.T., O.A.-W., M.G., E.M.P., A.M.M. and R.L.L. designed the clinical study, and collected and analyzed patient data. A.G.C., A.V.K., R.P.K. and S.A.A. designed, conducted and analyzed ChIP-sequencing experiments. B.S., F.E.G.-B. and A.M.M. assisted with RNA-sequencing, ERRBS and data analysis. B.S. mined TCGA data. L.L. and S.D.N. designed and performed peptide pull-down assay. B.H.D. assisted with animal hematopathology. A.M. and M.E.A. assisted with patient DNA re-sequencing. G.H., W.R.S., S.K. and P.B.S. designed and conducted ex vivo drug studies on primary AML samples. J.R.C. designed and oversaw small-molecule quantification by mass-spectroscopy. S.A.R., Y.K.L. and S.M. provided Dnmt3a cKO mice, S.M. contributed to study conception. O.A.-W. assisted with study design. O.A.G. and R.L.L. conceived the study, designed experiments, analyzed data and wrote the manuscript. All of the authors read, edited and approved the manuscript.

Corresponding authors

Correspondence to Siddhartha Mukherjee or Ross L Levine.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Tables 1–3, 7, 8 (PDF 15780 kb)

Supplementary Table 4

Differentially methylated cytosines (DMCs) in LSK cells from Dnmt3amut mice compared to wild-type controls, identified by MethyKit (XLSX 1235 kb)

Supplementary Table 5

Differentially expressed genes in LSK cells from Dnmt3amut mice compared to wild-type controls, identified by RNA-seq. (XLSX 58 kb)

Supplementary Table 6

Candidate DNMT3A interacting proteins identified by mass-spectroscopy following DNMT3A peptide pull-down. (XLSX 56 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Guryanova, O., Shank, K., Spitzer, B. et al. DNMT3A mutations promote anthracycline resistance in acute myeloid leukemia via impaired nucleosome remodeling. Nat Med 22, 1488–1495 (2016). https://doi.org/10.1038/nm.4210

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4210

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer