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:

MMSET regulates histone H4K20 methylation and 53BP1 accumulation at DNA damage sites

Abstract

p53-binding protein 1 (53BP1) is known to be an important mediator of the DNA damage response1, with dimethylation of histone H4 lysine 20 (H4K20me2) critical to the recruitment of 53BP1 to double-strand breaks (DSBs)2,3. However, it is not clear how 53BP1 is specifically targeted to the sites of DNA damage, as the overall level of H4K20me2 does not seem to increase following DNA damage. It has been proposed that DNA breaks may cause exposure of methylated H4K20 previously buried within the chromosome; however, experimental evidence for such a model is lacking. Here we found that H4K20 methylation actually increases locally upon the induction of DSBs and that methylation of H4K20 at DSBs is mediated by the histone methyltransferase MMSET (also known as NSD2 or WHSC1) in mammals. Downregulation of MMSET significantly decreases H4K20 methylation at DSBs and the subsequent accumulation of 53BP1. Furthermore, we found that the recruitment of MMSET to DSBs requires the γH2AX–MDC1 pathway; specifically, the interaction between the MDC1 BRCT domain and phosphorylated Ser 102 of MMSET. Thus, we propose that a pathway involving γH2AX–MDC1–MMSET regulates the induction of H4K20 methylation on histones around DSBs, which, in turn, facilitates 53BP1 recruitment.

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: Induction of H4K20 methylation and recruitment of MMSET at DSBs.
Figure 2: MMSET is required for H4K20me2/3 and 53BP1 accumulation at DSBs.
Figure 3: Recruitment of MMSET to DSBs requires the ATM–H2AX–MDC1 pathway.
Figure 4: Phosphorylation of MMSET is important for H4K20 methylation, 53BP1 recruitment and the DNA damage response.

Similar content being viewed by others

References

  1. FitzGerald, J. E., Grenon, M. & Lowndes, N. F. 53BP1: function and mechanisms of focal recruitment. Biochem. Soc. Trans. 37, 897–904 (2009)

    Article  CAS  Google Scholar 

  2. Botuyan, M. V. et al. Structural basis for the methylation state-specific recognition of histone H4-K20 by 53BP1 and Crb2 in DNA repair. Cell 127, 1361–1373 (2006)

    Article  CAS  Google Scholar 

  3. Sanders, S. L. et al. Methylation of histone H4 lysine 20 controls recruitment of Crb2 to sites of DNA damage. Cell 119, 603–614 (2004)

    Article  CAS  Google Scholar 

  4. Celeste, A. et al. Histone H2AX phosphorylation is dispensable for the initial recognition of DNA breaks. Nature Cell Biol. 5, 675–679 (2003)

    Article  CAS  Google Scholar 

  5. Bekker-Jensen, S., Lukas, C., Melander, F., Bartek, J. & Lukas, J. Dynamic assembly and sustained retention of 53BP1 at the sites of DNA damage are controlled by Mdc1/NFBD1. J. Cell Biol. 170, 201–211 (2005)

    Article  CAS  Google Scholar 

  6. Lou, Z. et al. MDC1 maintains genomic stability by participating in the amplification of ATM-dependent DNA damage signals. Mol. Cell 21, 187–200 (2006)

    Article  CAS  Google Scholar 

  7. Stewart, G. S., Wang, B., Bignell, C. R., Taylor, A. M. & Elledge, S. J. MDC1 is a mediator of the mammalian DNA damage checkpoint. Nature 421, 961–966 (2003)

    ADS  CAS  Google Scholar 

  8. Du, L. L., Nakamura, T. M. & Russell, P. Histone modification-dependent and -independent pathways for recruitment of checkpoint protein Crb2 to double-strand breaks. Genes Dev. 20, 1583–1596 (2006)

    Article  CAS  Google Scholar 

  9. Moynahan, M. E., Pierce, A. J. & Jasin, M. BRCA2 is required for homology-directed repair of chromosomal breaks. Mol. Cell 7, 263–272 (2001)

    Article  CAS  Google Scholar 

  10. Fang, J. et al. Purification and functional characterization of SET8, a nucleosomal histone H4-lysine 20-specific methyltransferase. Curr. Biol. 12, 1086–1099 (2002)

    Article  CAS  Google Scholar 

  11. Nishioka, K. et al. PR-Set7 is a nucleosome-specific methyltransferase that modifies lysine 20 of histone H4 and is associated with silent chromatin. Mol. Cell 9, 1201–1213 (2002)

    Article  CAS  Google Scholar 

  12. Schotta, G. et al. A silencing pathway to induce H3-K9 and H4-K20 trimethylation at constitutive heterochromatin. Genes Dev. 18, 1251–1262 (2004)

    Article  CAS  Google Scholar 

  13. Schotta, G. et al. A chromatin-wide transition to H4K20 monomethylation impairs genome integrity and programmed DNA rearrangements in the mouse. Genes Dev. 22, 2048–2061 (2008)

    Article  CAS  Google Scholar 

  14. Kim, J. Y. et al. Multiple-myeloma-related WHSC1/MMSET isoform RE-IIBP is a histone methyltransferase with transcriptional repression activity. Mol. Cell. Biol. 28, 2023–2034 (2008)

    Article  CAS  Google Scholar 

  15. Marango, J. et al. The MMSET protein is a histone methyltransferase with characteristics of a transcriptional corepressor. Blood 111, 3145–3154 (2008)

    Article  CAS  Google Scholar 

  16. Nimura, K. et al. A histone H3 lysine 36 trimethyltransferase links Nkx2-5 to Wolf-Hirschhorn syndrome. Nature 460, 287–291 (2009)

    Article  ADS  CAS  Google Scholar 

  17. Bergemann, A. D., Cole, F. & Hirschhorn, K. The etiology of Wolf-Hirschhorn syndrome. Trends Genet. 21, 188–195 (2005)

    Article  CAS  Google Scholar 

  18. Chesi, M. et al. The t(4;14) translocation in myeloma dysregulates both FGFR3 and a novel gene, MMSET, resulting in IgH/MMSET hybrid transcripts. Blood 92, 3025–3034 (1998)

    CAS  PubMed  Google Scholar 

  19. Stec, I. et al. WHSC1, a 90 kb SET domain-containing gene, expressed in early development and homologous to a Drosophila dysmorphy gene maps in the Wolf-Hirschhorn syndrome critical region and is fused to IgH in t(4;14) multiple myeloma. Hum. Mol. Genet. 7, 1071–1082 (1998)

    Article  CAS  Google Scholar 

  20. Wang, B., Matsuoka, S., Carpenter, P. B. & Elledge, S. J. 53BP1, a mediator of the DNA damage checkpoint. Science 298, 1435–1438 (2002)

    Article  ADS  CAS  Google Scholar 

  21. Huen, M. S. et al. RNF8 transduces the DNA-damage signal via histone ubiquitylation and checkpoint protein assembly. Cell 131, 901–914 (2007)

    Article  CAS  Google Scholar 

  22. Kolas, N. K. et al. Orchestration of the DNA-damage response by the RNF8 ubiquitin ligase. Science 318, 1637–1640 (2007)

    Article  ADS  CAS  Google Scholar 

  23. Mailand, N. et al. RNF8 ubiquitylates histones at DNA double-strand breaks and promotes assembly of repair proteins. Cell 131, 887–900 (2007)

    Article  CAS  Google Scholar 

  24. Stucki, M. et al. MDC1 directly binds phosphorylated histone H2AX to regulate cellular responses to DNA double-strand breaks. Cell 123, 1213–1226 (2005)

    Article  CAS  Google Scholar 

  25. Yu, X., Chini, C. C., He, M., Mer, G. & Chen, J. The BRCT domain is a phospho-protein binding domain. Science 302, 639–642 (2003)

    Article  ADS  CAS  Google Scholar 

  26. Manke, I. A., Lowery, D. M., Nguyen, A. & Yaffe, M. B. BRCT repeats as phosphopeptide-binding modules involved in protein targeting. Science 302, 636–639 (2003)

    Article  ADS  CAS  Google Scholar 

  27. Matsuoka, S. et al. ATM and ATR substrate analysis reveals extensive protein networks responsive to DNA damage. Science 316, 1160–1166 (2007)

    Article  ADS  CAS  Google Scholar 

  28. Lee, M. S., Edwards, R. A., Thede, G. L. & Glover, J. N. Structure of the BRCT repeat domain of MDC1 and its specificity for the free COOH-terminal end of the γ-H2AX histone tail. J. Biol. Chem. 280, 32053–32056 (2005)

    Article  CAS  Google Scholar 

  29. Kim, J. E., Chen, J. & Lou, Z. DBC1 is a negative regulator of SIRT1. Nature 451, 583–586 (2008)

    Article  ADS  CAS  Google Scholar 

  30. You, Z. et al. CtIP links DNA double-strand break sensing to resection. Mol. Cell 36, 954–969 (2009)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank M. Jasin for providing HeLa DR–GFP cells, and S. Baylin for providing MDA-MB-231 cells with inducible I-SceI expression. We thank M. Goldberg, X. Yu and M. Stucki for providing MDC1 and BRCA1 constructs. We thank M. Huen for providing 53BP1 expression constructs. We thank J. D. Licht for providing MMSET antibodies and B. Ho Park for providing MMSET shRNAs. This work was supported by the Richard Schulze Family Foundation and the NIH (CA130996 and CA151329; Z.L.), a grant from the American Cancer Society (IRG-58-010-52; Z.Y.) and a Siteman Career Award in Breast Cancer Research (Z.Y.).

Author information

Authors and Affiliations

Authors

Contributions

H.P. designed and performed the experiments; L.Z., K.L., Y.Q. and F.F. performed some experiments; M.C. and P.L.B. provided essential tools; L.W., Z.Y. and Z.L. designed the experiments and supervised the project.

Corresponding author

Correspondence to Zhenkun Lou.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Figures

The file contains Supplementary Figures 1-5 with legends and Supplementary Figures 1-4 for the raw data. (PDF 3329 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Pei, H., Zhang, L., Luo, K. et al. MMSET regulates histone H4K20 methylation and 53BP1 accumulation at DNA damage sites. Nature 470, 124–128 (2011). https://doi.org/10.1038/nature09658

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature09658

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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