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:

Histone H1 couples initiation and amplification of ubiquitin signalling after DNA damage

Abstract

DNA double-strand breaks (DSBs) are highly cytotoxic DNA lesions that trigger non-proteolytic ubiquitylation of adjacent chromatin areas to generate binding sites for DNA repair factors. This depends on the sequential actions of the E3 ubiquitin ligases RNF8 and RNF168 (refs 1, 2, 3, 4, 5, 6), and UBC13 (also known as UBE2N), an E2 ubiquitin-conjugating enzyme that specifically generates K63-linked ubiquitin chains7. Whereas RNF168 is known to catalyse ubiquitylation of H2A-type histones, leading to the recruitment of repair factors such as 53BP1 (refs 8, 9, 10), the critical substrates of RNF8 and K63-linked ubiquitylation remain elusive. Here we elucidate how RNF8 and UBC13 promote recruitment of RNF168 and downstream factors to DSB sites in human cells. We establish that UBC13-dependent K63-linked ubiquitylation at DSB sites is predominantly mediated by RNF8 but not RNF168, and that H1-type linker histones, but not core histones, represent major chromatin-associated targets of this modification. The RNF168 module (UDM1) recognizing RNF8-generated ubiquitylations11 is a high-affinity reader of K63-ubiquitylated H1, mechanistically explaining the essential roles of RNF8 and UBC13 in recruiting RNF168 to DSBs. Consistently, reduced expression or chromatin association of linker histones impair accumulation of K63-linked ubiquitin conjugates and repair factors at DSB-flanking chromatin. These results identify histone H1 as a key target of RNF8–UBC13 in DSB signalling and expand the concept of the histone code12,13 by showing that posttranslational modifications of linker histones can serve as important marks for recognition by factors involved in genome stability maintenance, and possibly beyond.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: RNF8 but not RNF168 is a primary mediator of UBC13-dependent protein recruitment to DSB-flanking chromatin.
Figure 2: H1-type linker histones are major chromatin-bound targets of RNF8–UBC13-dependent K63 ubiquitylation.
Figure 3: Histone H1 is required for K63-linked ubiquitylation and RNF168-dependent protein retention at DSB sites.
Figure 4: The RNF168 UDM1 domain is a high-affinity reader of K63-ubiquitylated H1.

Similar content being viewed by others

References

  1. 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 

  2. 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 

  3. 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 

  4. Doil, C. et al. RNF168 binds and amplifies ubiquitin conjugates on damaged chromosomes to allow accumulation of repair proteins. Cell 136, 435–446 (2009)

    Article  CAS  Google Scholar 

  5. Stewart, G. S. et al. The RIDDLE syndrome protein mediates a ubiquitin-dependent signaling cascade at sites of DNA damage. Cell 136, 420–434 (2009)

    Article  CAS  Google Scholar 

  6. Jackson, S. P. & Durocher, D. Regulation of DNA damage responses by ubiquitin and SUMO. Mol. Cell 49, 795–807 (2013)

    Article  CAS  Google Scholar 

  7. Hofmann, R. M. & Pickart, C. M. Noncanonical MMS2-encoded ubiquitin-conjugating enzyme functions in assembly of novel polyubiquitin chains for DNA repair. Cell 96, 645–653 (1999)

    CAS  PubMed  Google Scholar 

  8. Mattiroli, F. et al. RNF168 ubiquitinates K13-15 on H2A/H2AX to drive DNA damage signaling. Cell 150, 1182–1195 (2012)

    Article  CAS  Google Scholar 

  9. Fradet-Turcotte, A. et al. 53BP1 is a reader of the DNA-damage-induced H2A Lys 15 ubiquitin mark. Nature 499, 50–54 (2013)

    Article  ADS  CAS  Google Scholar 

  10. Gatti, M. et al. A novel ubiquitin mark at the N-terminal tail of histone H2As targeted by RNF168 ubiquitin ligase. Cell Cycle 11, 2538–2544 (2012)

    Article  CAS  Google Scholar 

  11. Panier, S. et al. Tandem protein interaction modules organize the ubiquitin-dependent response to DNA double-strand breaks. Mol. Cell 47, 383–395 (2012)

    Article  CAS  Google Scholar 

  12. Kouzarides, T. Chromatin modifications and their function. Cell 128, 693–705 (2007)

    Article  CAS  Google Scholar 

  13. Jenuwein, T. & Allis, C. D. Translating the histone code. Science 293, 1074–1080 (2001)

    Article  CAS  Google Scholar 

  14. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013)

    Article  ADS  CAS  Google Scholar 

  15. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, 823–826 (2013)

    Article  ADS  CAS  Google Scholar 

  16. Bekker-Jensen, S. et al. HERC2 coordinates ubiquitin-dependent assembly of DNA repair factors on damaged chromosomes. Nature Cell Biol. 12, 80–86,–1–12 (2010)

    Google Scholar 

  17. Huen, M. S. et al. Noncanonical E2 variant-independent function of UBC13 in promoting checkpoint protein assembly. Mol. Cell. Biol. 28, 6104–6112 (2008)

    Article  CAS  Google Scholar 

  18. Sims, J. J. et al. Polyubiquitin-sensor proteins reveal localization and linkage-type dependence of cellular ubiquitin signaling. Nature Methods 9, 303–309 (2012)

    Article  CAS  Google Scholar 

  19. van Wijk, S. J. et al. Fluorescence-based sensors to monitor localization and functions of linear and K63-linked ubiquitin chains in cells. Mol. Cell 47, 797–809 (2012)

    Article  CAS  Google Scholar 

  20. Wagner, S. A. et al. A proteome-wide, quantitative survey of in vivo ubiquitylation sites reveals widespread regulatory roles. Mol. Cell. Proteomics 10, M111.013284 (2011)

    Article  Google Scholar 

  21. Kim, W. et al. Systematic and quantitative assessment of the ubiquitin-modified proteome. Mol. Cell 44, 325–340 (2011)

    Article  CAS  Google Scholar 

  22. Petroski, M. D. et al. Substrate modification with lysine 63-linked ubiquitin chains through the UBC13-UEV1A ubiquitin-conjugating enzyme. J. Biol. Chem. 282, 29936–29945 (2007)

    Article  CAS  Google Scholar 

  23. Christensen, D. E., Brzovic, P. S. & Klevit, R. E. E2-BRCA1 RING interactions dictate synthesis of mono- or specific polyubiquitin chain linkages. Nature Struct. Mol. Biol. 14, 941–948 (2007)

    Article  CAS  Google Scholar 

  24. Windheim, M., Peggie, M. & Cohen, P. Two different classes of E2 ubiquitin-conjugating enzymes are required for the mono-ubiquitination of proteins and elongation by polyubiquitin chains with a specific topology. Biochem. J. 409, 723–729 (2008)

    Article  CAS  Google Scholar 

  25. Catez, F., Ueda, T. & Bustin, M. Determinants of histone H1 mobility and chromatin binding in living cells. Nature Struct. Mol. Biol. 13, 305–310 (2006)

    Article  CAS  Google Scholar 

  26. Murga, M. et al. Global chromatin compaction limits the strength of the DNA damage response. J. Cell Biol. 178, 1101–1108 (2007)

    Article  CAS  Google Scholar 

  27. Pinato, S., Gatti, M., Scandiuzzi, C., Confalonieri, S. & Penengo, L. UMI, a novel RNF168 ubiquitin binding domain involved in the DNA damage signaling pathway. Mol. Cell. Biol. 31, 118–126 (2011)

    Article  CAS  Google Scholar 

  28. Harshman, S. W., Young, N. L., Parthun, M. R. & Freitas, M. A. H1 histones: current perspectives and challenges. Nucleic Acids Res. 41, 9593–9609 (2013)

    Article  CAS  Google Scholar 

  29. Gatti, M. et al. RNF168 promotes noncanonical K27 ubiquitination to signal DNA damage. Cell Reports 10, 226–238 (2015)

    Article  CAS  Google Scholar 

  30. Povlsen, L. K. et al. Systems-wide analysis of ubiquitylation dynamics reveals a key role for PAF15 ubiquitylation in DNA-damage bypass. Nature Cell Biol. 14, 1089–1098 (2012)

    Article  CAS  Google Scholar 

  31. Danielsen, J. M. et al. Mass spectrometric analysis of lysine ubiquitylation reveals promiscuity at site level. Mol. Cell. Proteomics 10, M110.003590 (2011)

    Article  Google Scholar 

  32. Poulsen, M., Lukas, C., Lukas, J., Bekker-Jensen, S. & Mailand, N. Human RNF169 is a negative regulator of the ubiquitin-dependent response to DNA double-strand breaks. J. Cell Biol. 197, 189–199 (2012)

    Article  CAS  Google Scholar 

  33. 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 

  34. Hernández-Muñoz, I. et al. Stable X chromosome inactivation involves the PRC1 Polycomb complex and requires histone MACROH2A1 and the CULLIN3/SPOP ubiquitin E3 ligase. Proc. Natl Acad. Sci. USA 102, 7635–7640 (2005)

    Article  ADS  Google Scholar 

  35. Ekkebus, R. et al. On terminal alkynes that can react with active-site cysteine nucleophiles in proteases. J. Am. Chem. Soc. 135, 2867–2870 (2013)

    Article  CAS  Google Scholar 

  36. Ong, S. E. et al. Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Mol. Cell. Proteomics 1, 376–386 (2002)

    Article  CAS  Google Scholar 

  37. Jensen, O. N., Wilm, M., Shevchenko, A. & Mann, M. Sample preparation methods for mass spectrometric peptide mapping directly from 2-DE gels. Methods Mol. Biol. 112, 513–530 (1999)

    CAS  PubMed  Google Scholar 

  38. Cox, J. & Mann, M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nature Biotechnol. 26, 1367–1372 (2008)

    Article  CAS  Google Scholar 

  39. Elias, J. E. & Gygi, S. P. Target-decoy search strategy for increased confidence in large-scale protein identifications by mass spectrometry. Nature Methods 4, 207–214 (2007)

    Article  CAS  Google Scholar 

  40. Schaefer, M. H. et al. HIPPIE: Integrating protein interaction networks with experiment based quality scores. PLoS ONE 7, e31826 (2012)

    Article  ADS  CAS  Google Scholar 

  41. Cline, M. S. et al. Integration of biological networks and gene expression data using Cytoscape. Nature Protocols 2, 2366–2382 (2007)

    Article  ADS  CAS  Google Scholar 

Download references

Acknowledgements

We thank D. Durocher and M. Bianchi for providing reagents and J. Lukas for helpful discussions. This work was supported by grants from the Novo Nordisk Foundation (grants NNF14CC0001 and NNF12OC0002114), European Research Council, Nederlandse Organisatie voor Wetenschappelijk Onderzoek- Chemische Wetenschappen (NWO-CW), The Danish Cancer Society, and The Danish Council for Independent Research.

Author information

Authors and Affiliations

Authors

Contributions

T.T. initiated the project, designed and performed cell biological and biochemical experiments, and analysed data; A.R. and S.H. performed cell biological and biochemical experiments and analysed data; T.W. generated UBC13-knockout cells, designed and performed mass spectrometry experiments and analysed the data; M.U. performed in vitro ubiquitylation assays with purified nucleosomes; B.V. helped T.T. with biochemical experiments; T.N. analysed mass spectrometry data; T.K.S. supervised M.U.; C.C. supervised T.W. and T.N., designed mass spectrometry experiments and analysed the data; S.B.-J. performed and designed experiments and analysed data; N.M. conceived and supervised the project, designed experiments, analysed data and wrote the manuscript with input from the other authors.

Corresponding author

Correspondence to Niels Mailand.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Protein recruitment to DSB sites in UBC13-knockout cells.

a, b, Representative images of HCT116 wild-type (WT) or UBC13-knockout (KO) cells exposed to IR. Where indicated, cells were transfected with HA–RNF8 plasmid before IR (n = 2 experiments). c, Constructs used in Fig. 1d and their ability to restore IR-induced 53BP1 foci in UBC13-knockout cells. d, Representative images of HCT116 UBC13-knockout cells transfected with non-targeting control (CTRL) or RNF8 siRNAs and subsequently with plasmids encoding UBC13 or siRNA-resistant UBC13–RNF8 fusion constructs (c) (n = 2). e, Representative images of UBC13-knockout cells transfected with plasmids encoding wild-type or catalytically inactive (CI) HA–RNF168 (n = 2). Expression of HA–RNF168 wild type restores IR-induced 53BP1 foci formation (arrows). Scale bars, 10 μm.

Extended Data Figure 2 RNF8- and UBC13-dependent K63-linked ubiquitylation at DSB sites.

ac, Representative images of U2OS cells stably expressing GFP–K63-Super-UIM, transfected with siRNAs where indicated, and exposed to laser micro-irradiation or IR (n = 3). d, Representative images of U2OS cells stably expressing GFP- and nuclear localization signal (NLS)-tagged Vps27-UIM (with high affinity for binding to K63-linked ubiquitin18) transfected with the indicated siRNAs and exposed to laser micro-irradiation (n = 2). e, Loss of RNF8 or UBC13 has no impact on 53BP1 abundance. Immunoblot analysis of whole-cell extracts (WCEs) of U2OS cells transfected with indicated siRNAs. f, RNF8, but not RNF168, is modified by K63-linked ubiquitin chains. K63-Super-UIM pull-downs from U2OS cells transfected with empty vector (−), HA-tagged RNF8 (wild type (WT) or catalytically inactive (CI)) or HA–RNF168 plasmids were immunoblotted (IB) with indicated antibodies. Scale bars, 10 μm. e, f, Uncropped blots are shown in Supplementary Fig. 1.

Extended Data Figure 3 Experimental replicates of SILAC-based quantification of di-glycine-containing peptides and K63-Super-UIM pull-downs from wild-type and UBC13-knockout cells.

a, Schematic outline of SILAC-based mass spectrometry approach to quantify di-glycine-containing peptides in HCT116 wild-type (WT) and UBC13-knockout (KO) cells. b, c, Proportional Venn diagrams showing overlap between all identified di-glycine-containing peptides (b) and those with a SILAC ratio (UBC13-knockout/wild-type cells) <0.5 (c) in two independent experiments (Exp.) performed as shown in a (Supplementary Table 1). d, Scatter plot showing correlation between SILAC ratios of di-glycine-containing peptides. The Pearson’s correlation coefficient (R) is indicated. e, Schematic outline of SILAC-based mass spectrometry approach to identify UBC13-dependent K63-ubiquitylation targets in unperturbed HCT116 wild-type and UBC13-knockout cells. f, g, Proportional Venn diagrams showing overlap between all proteins identified in K63-Super-UIM pull-downs (f) and those with a SILAC ratio (UBC13-knockout/wild-type cells) <0.5 (g) in two independent experiments performed as shown in e (Supplementary Table 2). h, Scatter plot showing correlation between SILAC ratios of proteins identified in two experiments. The Pearson’s correlation coefficient (R) is indicated.

Extended Data Figure 4 Analysis of UBC13-dependent K63-ubiquitylated proteins in unperturbed cells and in response to DNA damage.

a, K63 linkages from extracts of HCT116 wild-type (WT) and UBC13-knockout (KO) cells were enriched by K63-Super-UIM pull-down (Supplementary Table 2). Interaction network shows proteins enriched at least twofold in wild-type cells. Proteins involved in UBC13-dependent activation of NF-κB signalling are highlighted in red. b, Functional annotation of potential UBC13-dependent K63-ubiquitylated proteins (a), showing enriched Gene Ontology (GO) biological process terms. c, Schematic outline of SILAC-based mass spectrometry approach to identify targets of K63 ubiquitylation in response to IR-induced DSBs. d, SILAC ratios of selected proteins from U2OS cells treated as in c. Data from a representative experiment are shown (n = 3).

Extended Data Figure 5 DSB-induced K63 ubiquitylation of H1-type linker histones.

a, Analysis of K63-linked ubiquitylation of histone H1.2 in RPE1 cells growing exponentially or kept quiescent by serum starvation. b, c, K63-Super-UIM pull-downs from U2OS cells exposed or not to IR were immunoblotted (IB) with the indicated antibodies. d, U2OS cells or U2OS cells stably expressing Strep–HA–ubiquitin (U2OS/Strep–Ub) were transfected with the indicated siRNAs and exposed or not to IR. Whole-cell extracts (WCEs) and Strep–ubiquitin-conjugated proteins immobilized on Strep-Tactin beads under denaturing conditions were analysed by immunoblotting. e, Proteins interacting with endogenous RNF8. U2OS cells stably expressing RNF8 shRNA in a doxycycline (DOX)-inducible manner1 was grown in light (L) or heavy (H) SILAC medium. Cells growing in light medium were induced to express RNF8 shRNA by treatment with DOX. Both cultures were then exposed to IR and processed for immunoprecipitation with RNF8 antibody. Bound proteins were analysed by mass spectrometry. Proteins displaying the highest H/L SILAC ratios are listed. ad, The migration of molecular weight markers (kDa) is indicated on the left. Uncropped blots are shown in Supplementary Fig. 1.

Extended Data Figure 6 Knockdown of H1-type histones impairs accumulation of K63-linked ubiquitin conjugates, RNF168 and BRCA1 at DSB sites.

a, Immunoblot analysis of U2OS cells transfected sequentially with plasmids encoding GFP-tagged histone H1 isoforms (H1.0–H1.5) and the indicated siRNAs. Knockdown efficiency of the siRNAs (#1, #4, #5 and #9) used in the pan-H1 siRNA cocktail to reduce global H1 expression level is highlighted in red boxes. b, Immunoblot analysis of U2OS cells transfected with H1 siRNA cocktail (a). cg, Representative images of siRNA-transfected U2OS/GFP–K63-Super UIM (c) or U2OS cells (dg) exposed to IR or laser micro-irradiation (n = 2). h, Analysis of IR-induced γ-H2AX ubiquitylation (Ub) by RNF168 (marked by arrow) in chromatin fractions of U2OS cells transfected with indicated siRNAs. Scale bars, 10 μm. a, b, h, Uncropped blots are shown in Supplementary Fig. 1.

Extended Data Figure 7 HMGB1 overexpression impairs the RNF8/RNF168-dependent signalling response at DSB sites at the level of K63 ubiquitylation and RNF168 recruitment.

a, b, Representative images of U2OS cells co-transfected with constructs encoding HMGB1–GFP and wild-type (WT) or catalytically inactive (CI) HA–RNF168 and exposed to IR (n = 3). Arrows indicate cells expressing HA–RNF168 CI, in which 53BP1 foci formation is not restored. c, Representative images of U2OS/GFP–K63-Super-UIM cells transfected with Flag–HMGB1 construct and subjected to laser micro-irradiation (n = 3). Flag–HMGB1-expressing cells show reduced K63 ubiquitylation at DSB sites (indicated by arrows). Scale bars, 10 μm.

Extended Data Figure 8 Interaction of RNF168 UDM1 with K63-ubiquitylated H1.

a, Immunoblot analysis of immobilized recombinant RNF168 UDM1 or UDM2 incubated with K63 linked di-ubiquitin (Ub2) in the presence of increasing salt concentrations (75 mM, 150 mM and 250 mM KCl, respectively). b, Binding of immobilized recombinant Strep–UDM2 or empty Strep-Tactin beads to indicated di-ubiquitin (Ub2) linkages was analysed by immunoblotting. c, Biotinylated peptides corresponding to the LRM1 and LRM2 motifs in human RNF168 were analysed for binding to recombinant H2A or H1.0 in vitro by Streptavidin pull-down followed by SDS–PAGE and Colloidal Blue staining. The migration of molecular weight markers (kDa) is indicated on the left. d, Sequence of the UDM1 region in human RNF168, showing the location of the LRM1, UMI and MIU1 motifs. Acidic amino acids are highlighted in red. The sequence corresponding to the LRM1 peptide (c) and mutations introduced to generate UDM1 *UMI and *MIU1 (f) are indicated. e, f, Pull-down assays of Strep-tagged UDM1 and UDM2 constructs expressed in U2OS cells. ac, e, f, Uncropped blots are shown in Supplementary Fig. 1.

Extended Data Figure 9 RNF168 UDM1 recognizes ubiquitylated forms of H1 but not core histones.

a, Pull-downs of Strep-tagged RNF168 UDM1 expressed in U2OS cells were immunoblotted (IB) with antibodies to indicated histones. b, Localization pattern of GFP-tagged UDM1 expressed in U2OS cells. Scale bar, 10 μm. c, Venn diagram showing overlap between proteins displaying increased K63-linked ubiquitylation after IR (SILAC ratio (IR/mock) >1.5) and proteins showing potential interaction with overexpressed GFP–UDM1 (SILAC ratio (GFP–UDM1/mock) >1.5). Only one protein, histone H1x, was common to both of these subsets of cellular proteins. a, Uncropped blots are shown in Supplementary Fig. 1.

Extended Data Figure 10 Impact of DSBs on H1 chromatin association.

a, FRAP analysis of U2OS cells stably expressing GFP–H1.2 and exposed or not to IR (10 Gy). Individual data points represent mean values from ten independent measurements and error bars represent twice the s.d. b, U2OS cells left untreated or exposed to IR were lysed in EBC buffer. Soluble and resolubilized, EBC-insoluble fractions were incubated with recombinant K63-Super-UIM and washed thoroughly. Bound material and input fractions were analysed by immunoblotting with indicated antibodies. b, Uncropped blots are shown in Supplementary Fig. 1.

Supplementary information

Supplementary Figure 1

This file contains the uncropped scans of immunoblot data with size marker indications. (PDF 5793 kb)

Supplementary Table 1

This table contains the Mass spectrometry data for SILAC-based quantification of di-glycine containing peptides isolated from HCT116 WT and Ubc13 KO cells. See Extended Data Fig. 3a for experimental set-up. (XLSX 1569 kb)

Supplementary Table 2

This table contains the Quantitative mass spectrometry data for proteins identified in K63-Super-UIM pull-downs from HCT116 WT and Ubc13 KO cells. See Extended Data Fig. 3e for experimental set-up. (XLSX 780 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Thorslund, T., Ripplinger, A., Hoffmann, S. et al. Histone H1 couples initiation and amplification of ubiquitin signalling after DNA damage. Nature 527, 389–393 (2015). https://doi.org/10.1038/nature15401

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature15401

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