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.

  • Brief Communication
  • Published:

Mechanism of histone H2B monoubiquitination by Bre1

Abstract

Monoubiquitination of histone H2B-K120/123 plays several roles in regulating transcription, DNA replication and the DNA damage response. The structure of a nucleosome in complex with the dimeric RING E3 ligase Bre1 reveals that one RING domain binds to the nucleosome acidic patch, where it can position the E2 ubiquitin conjugating enzyme Rad6, while the other RING domain contacts the DNA. Comparisons with H2A-specific E3 ligases suggest a general mechanism of tuning histone specificity via the non-E2-binding RING domain.

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: Cryo-EM structures of Bre1 dimer bound to nucleosome reveal several conformations.
Fig. 2: Bre1 orients Rad6 to ubiquitinate histone H2B-K120.

Similar content being viewed by others

Data availability

The maps are available in the Electron Microscopy Data Bank (EMDB) database under accession no. EMD-41016 (state 1), EMD-41015 (state 2) and EMD-41011 (state 3). The atomic models are available in the Protein Data Bank database: PDB 8T3Y (state 1), PDB 8T3W(state 2) and PDB 8T3T (state 3). Source data are provided with this paper.

References

  1. Bonnet, J., Devys, D. & Tora, L. Histone H2B ubiquitination: signaling not scrapping. Drug Discov. Today Technol. 12, e19–e27 (2014).

    Article  PubMed  Google Scholar 

  2. Mattiroli, F. & Penengo, L. Histone ubiquitination: an integrative signaling platform in genome stability. Trends Genet. 37, 566–581 (2021).

    Article  CAS  PubMed  Google Scholar 

  3. Weake, V. M. & Workman, J. L. Histone ubiquitination: triggering gene activity. Mol. Cell 29, 653–663 (2008).

    Article  CAS  PubMed  Google Scholar 

  4. Fleming, A. B., Kao, C. F., Hillyer, C., Pikaart, M. & Osley, M. A. H2B ubiquitylation plays a role in nucleosome dynamics during transcription elongation. Mol. Cell 31, 57–66 (2008).

    Article  CAS  PubMed  Google Scholar 

  5. Pavri, R. et al. Histone H2B monoubiquitination functions cooperatively with FACT to regulate elongation by RNA polymerase II. Cell 125, 703–717 (2006).

    Article  CAS  PubMed  Google Scholar 

  6. Hwang, W. W. et al. A conserved RING finger protein required for histone H2B monoubiquitination and cell size control. Mol. Cell 11, 261–266 (2003).

    Article  CAS  PubMed  Google Scholar 

  7. Robzyk, K., Recht, J. & Osley, M. A. Rad6-dependent ubiquitination of histone H2B in yeast. Science 287, 501–504 (2000).

    Article  CAS  PubMed  Google Scholar 

  8. Kim, J., Hake, S. B. & Roeder, R. G. The human homolog of yeast BRE1 functions as a transcriptional coactivator through direct activator interactions. Mol. Cell 20, 759–770 (2005).

    Article  CAS  Google Scholar 

  9. Kim, J. et al. RAD6-Mediated transcription-coupled H2B ubiquitylation directly stimulates H3K4 methylation in human cells. Cell 137, 459–471 (2009).

    Article  CAS  PubMed Central  Google Scholar 

  10. Sethi, G., Shanmugam, M. K., Arfuso, F. & Kumar, A. P. Role of RNF20 in cancer development and progression—a comprehensive review. Biosci. Rep. 38, BSR20171287 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Kumar, P. & Wolberger, C. Structure of the yeast Bre1 RING domain. Proteins 83, 1185–1190 (2015).

    Article  CAS  PubMed Central  Google Scholar 

  12. Turco, E., Gallego, L. D., Schneider, M. & Kohler, A. Monoubiquitination of histone H2B is intrinsic to the Bre1 RING domain-Rad6 interaction and augmented by a second Rad6-binding site on Bre1. J. Biol. Chem. 290, 5298–5310 (2015).

    Article  CAS  PubMed  Google Scholar 

  13. Kim, J. & Roeder, R. G. Direct Bre1–Paf1 complex interactions and RING finger-independent Bre1–Rad6 interactions mediate histone H2B ubiquitylation in yeast. J. Biol. Chem. 284, 20582–20592 (2009).

    Article  CAS  PubMed Central  Google Scholar 

  14. Gallego, L. D. et al. Structural mechanism for the recognition and ubiquitination of a single nucleosome residue by Rad6–Bre1. Proc. Natl Acad. Sci. USA 113, 10553–10558 (2016).

    Article  CAS  PubMed Central  Google Scholar 

  15. McGinty, R. K. & Tan, S. Principles of nucleosome recognition by chromatin factors and enzymes. Curr. Opin. Struct. Biol. 71, 16–26 (2021).

    Article  CAS  PubMed Central  Google Scholar 

  16. Gundogdu, M. & Walden, H. Structural basis of generic versus specific E2-RING E3 interactions in protein ubiquitination. Protein Sci. 28, 1758–1770 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  17. Pan, M. et al. Structural insights into Ubr1-mediated N-degron polyubiquitination. Nature 600, 334–338 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Hu, Q. et al. Mechanisms of BRCA1-BARD1 nucleosome recognition and ubiquitylation. Nature 596, 438–443 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Witus, S. R. et al. BRCA1/BARD1 site-specific ubiquitylation of nucleosomal H2A is directed by BARD1. Nat. Struct. Mol. Biol. 28, 268–277 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Shukla, P. K. et al. Structure and functional determinants of Rad6-Bre1 subunits in the histone H2B ubiquitin-conjugating complex.Nucleic Acids Res. 51, 2117–2136 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. McGinty, R. K., Henrici, R. C. & Tan, S. Crystal structure of the PRC1 ubiquitylation module bound to the nucleosome. Nature 514, 591–596 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Worden, E. J., Hoffmann, N. A., Hicks, C. W. & Wolberger, C. Mechanism of cross-talk between H2B ubiquitination and H3 methylation by Dot1L. Cell 176, 1490–1501 e12 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Dyer, P. N. et al. Reconstitution of nucleosome core particles from recombinant histones and DNA.Methods Enzymol 375, 23–44 (2004).

    Article  CAS  PubMed  Google Scholar 

  24. Punjani, A., Rubinstein, J. L., Fleet, D. J. & Brubaker, M. A. cryoSPARC: algorithms for rapid unsupervised cryo-EM structure determination. Nat. Methods 14, 290–296 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Goddard, T. D. et al. UCSF ChimeraX: meeting modern challenges in visualization and analysis. Protein Sci. 27, 14–25 (2018).

    Article  CAS  PubMed  Google Scholar 

  26. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot. Acta Crystallogr D. Biol. Crystallogr 66, 486–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Afonine, P. V. et al. Real-space refinement in PHENIX for cryo-EM and crystallography. Acta Crystallogr D. Struct. Biol. 74, 531–544 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Liebschner, D. et al. Macromolecular structure determination using X-rays, neutrons and electrons: recent developments in Phenix. Acta Crystallogr D. Struct. Biol. 75, 861–877 (2019).

    Article  CAS  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Johns Hopkins colleagues C. Liu and E. Twomey for advice on Cryo-EM data collection and processing and X. Zhang for valuable suggestions on constructing plasmids. Supported in part by the National Cancer Institute’s National Cryo-EM Facility at the Frederick National Laboratory for Cancer Research under contract 75N91019D00024. This work was supported by National Institute of General Medical Sciences grant GM130393 (C.W.).

Author information

Authors and Affiliations

Authors

Contributions

C.W. and F.Z. conceived and designed the study. F.Z. and C.W.H. performed experiments and processed data. F.Z. carried out modeling and map interpretation with assistance from C.W. F.Z., C.W.H. and C.W. wrote the manuscript. C.W. supervised the study.

Corresponding author

Correspondence to Cynthia Wolberger.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Structural & Molecular Biology thanks Uhn-Soo Cho and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Sara Osman and Carolina Perdigoto were the primary editors on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team. Peer reviewer reports are available.

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 Cryo-EM processing workflow.

The red dashed line indicates the mask generated for use in 3D classification. Three states of Bre1–nucleosome complex are identified and refined respectively.

Extended Data Fig. 2 Cryo-EM validation and example density.

a–c, Local resolution estimation and gold-standard Fourier shell correlation (GSFSC) curves at a FSC cutoff of 0.143 for each state of Bre1–nucleosome complex. d, Representative cryo-EM density maps for the histones, DNA, and Bre1.

Extended Data Fig. 3 Comparison of enzymatic activity of fused and separated Bre1 and Rad6 enzyme.

In vitro nucleosome H2B ubiquitination assay was performed with fused or separated Bre1 and Rad6 enzyme. Samples were quenched by SDS-loading buffer at indicated timepoints and subjected to immunoblotting. Data is representative of 2 independent experiments.

Source data

Extended Data Fig. 4 Electrostatic surface representation view of Bre1 dimer (PDB:4R7E).

Bre1 is shown in a cartoon representation with basic residues highlighted as sticks.

Extended Data Fig. 5 Multiple conformations of the Bre1–nucleosome complex.

a–c, Cryo-EM maps of the Bre1–nucleosome complex with an atomic model fitted to states 1, 2 and 3. d, Superimpose of Bre1–nucleosome complex maps comparing states 1, 2 and 3. White arrows show the changes between each conformational state.

Extended Data Fig. 6 RNF20/40 mutation sites mapped to RING domain and preceded α-helix in cancer genomics.

a, Sequence alignment of coiled coil and RING domains of Bre1, RNF20 and RNF40. Black triangle indicates the mutation sites of RNF20 and RNF40 in cancer genomics. b, Complex structure model of Bre1 RING-Rad6-nucleosome. Bre1 residues corresponding to RNF20/40 mutated sites were shown as sticks with dot sphere.

Extended Data Fig. 7 Canonical interface between E3 RING and E2.

a, Superposition of multiple E2-E3 structures. E2-E3 complex structures of RNF4-UbcH5a (PDB: 4AP4), Ubr1-Rad6 (PDB: 7MEX), Ring1b-UbcH5c (PDB: 4R8P), BRCA1-UbcH5c (PDB: 7LYB) and TRIM25-UbcH5 (PDB: 5FER) are aligned together. b, The modeled structure of Bre1 RING-Rad6. c, Residues located in modeled Bre1A RING-Rad6 interface. The side chains of residues located in the interface are shown as sticks.

Extended Data Fig. 8 Superposition of Bre1–nucleosome complex (state 2 and 1) and Ubr1 RING-Rad6-ubiquitin (PDB: 7MEX).

a, c, Ubr1 RING domain is aligned to Bre1-A RING domain in state 2 (a) and state 1 (c). b, d, Close-up view of Rad6 active site C88, ubiquitin G76, and H2B K120 in state 2 (b) and state 1 (d). The H2B K120 rotamer with the shortest distance from Rad6 C88 and ubiquitin G76 is shown here.

Extended Data Fig. 9 Structural alignment of Bre1 RBD-Rad6 complex and Bre1 RING-Rad6-Ub-nucleosome complex (model of state 3).

a, b, Rad6 in Bre1 RBD-Rad6 complex is aligned to the Rad6 in Bre1 RING-Rad6-Ub-nucleosome complex (model of state 3).

Supplementary information

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhao, F., Hicks, C.W. & Wolberger, C. Mechanism of histone H2B monoubiquitination by Bre1. Nat Struct Mol Biol 30, 1623–1627 (2023). https://doi.org/10.1038/s41594-023-01137-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-023-01137-x

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