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.

  • Article
  • Published:

Basis of the H2AK119 specificity of the Polycomb repressive deubiquitinase

Abstract

Repression of gene expression by protein complexes of the Polycomb group is a fundamental mechanism that governs embryonic development and cell-type specification1,2,3. The Polycomb repressive deubiquitinase (PR-DUB) complex removes the ubiquitin moiety from monoubiquitinated histone H2A K119 (H2AK119ub1) on the nucleosome4, counteracting the ubiquitin E3 ligase activity of Polycomb repressive complex 1 (PRC1)5 to facilitate the correct silencing of genes by Polycomb proteins and safeguard active genes from inadvertent silencing by PRC1 (refs. 6,7,8,9). The intricate biological function of PR-DUB requires accurate targeting of H2AK119ub1, but PR-DUB can deubiquitinate monoubiquitinated free histones and peptide substrates indiscriminately; the basis for its exquisite nucleosome-dependent substrate specificity therefore remains unclear. Here we report the cryo-electron microscopy structure of human PR-DUB, composed of BAP1 and ASXL1, in complex with the chromatosome. We find that ASXL1 directs the binding of the positively charged C-terminal extension of BAP1 to nucleosomal DNA and histones H3–H4 near the dyad, an addition to its role in forming the ubiquitin-binding cleft. Furthermore, a conserved loop segment of the catalytic domain of BAP1 is situated near the H2A–H2B acidic patch. This distinct nucleosome-binding mode displaces the C-terminal tail of H2A from the nucleosome surface, and endows PR-DUB with the specificity for H2AK119ub1.

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: Structure of PR-DUB bound to the H2AK119ub1 chromatosome.
Fig. 2: Structure and function of the BAP1 CTE.
Fig. 3: Nucleosome binding is key to H2AK119ub1 deubiquitination.
Fig. 4: Conformational change of the H2A C-terminal tail and the role of H1 in PR-DUB activity.

Similar content being viewed by others

Data availability

The cryo-EM density maps have been deposited in the Electron Microscopy Data Bank (EMDB) with accession numbers EMD-34431 and EMD-34432 for the chromatosomal and nucleosomal complexes, respectively. Constituent and consensus maps for the chromatosomal complex have been deposited with accession numbers EMD-35179, EMD-35180, EMD-35181 and EMD-35182. The atomic coordinates for the structure of PR-DUB bound to the chromatosome have been deposited in the PDB with the accession code 8H1T. The coordinates for the structures of Drosophila Caly in complex with DEUBAD of ASX (PDB ID: 6HGC), H2AK15-ubiquitnated NCP in complex with the BRCA1–BARD complex (PDB ID: 7E8I), the H2BK120-ubiqutinated NCP in complex with the SAGA DUB module (PDB ID: 4ZUX), the UCH-L5 complex with DEUBAD of RPN13 and ubiquitin (PDB ID: 4UEL) and the chromatosome with linker histone H1.4 (PDB ID: 7K5Y) were downloaded from the RCSB Protein Data Bank (https://www.rcsb.org). The RNA-seq data have been deposited in the Genome Sequence Archive at the National Genomics Data Center, China National Center for Bioinformation–Beijing Institute of Genomics, Chinese Academy of Sciences (GSA: CRA006013) and are publicly accessible at https://ngdc.cncb.ac.cn/gsa. Two public RNA-seq datasets with the accession numbers GSE162739 and GSE161995 were used in this study. The list of differentially expressed genes (DEGs) defined in a previous study7 was downloaded from ref. 7 (https://www.sciencedirect.com/science/article/pii/S1097276521005001?via%3Dihub#app2), with the original file name of 1-s2.0-S1097276521005001-mmc3.xlsx. The list of DEGs defined in a previous study8. was downloaded from the Gene Expression Omnibus (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE161995), with the original file name of GSE161995_BAP1ff_cnRNA_seq_DESeq2_Table_GEO.txt. A threshold of Padj < 0.05 and fold change > 1.5 was used to extract the DEGs. Source data are provided with this paper.

References

  1. Simon, J. A. & Kingston, R. E. Mechanisms of polycomb gene silencing: knowns and unknowns. Nat. Rev. Mol. Cell Biol. 10, 697–708 (2009).

    Article  CAS  PubMed  Google Scholar 

  2. Schuettengruber, B., Bourbon, H. M., Di Croce, L. & Cavalli, G. Genome regulation by Polycomb and Trithorax: 70 years and counting. Cell 171, 34–57 (2017).

    Article  CAS  PubMed  Google Scholar 

  3. Blackledge, N. P. & Klose, R. J. The molecular principles of gene regulation by Polycomb repressive complexes. Nat. Rev. Mol. Cell Biol. 22, 815–833 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Scheuermann, J. C. et al. Histone H2A deubiquitinase activity of the Polycomb repressive complex PR-DUB. Nature 465, 243–247 (2010).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  5. Wang, H. et al. Role of histone H2A ubiquitination in Polycomb silencing. Nature 431, 873–878 (2004).

    Article  ADS  CAS  PubMed  Google Scholar 

  6. Campagne, A. et al. BAP1 complex promotes transcription by opposing PRC1-mediated H2A ubiquitylation. Nat. Commun. 10, 348 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  7. Conway, E. et al. BAP1 enhances Polycomb repression by counteracting widespread H2AK119ub1 deposition and chromatin condensation. Mol. Cell 81, 3526–3541 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Fursova, N. A. et al. BAP1 constrains pervasive H2AK119ub1 to control the transcriptional potential of the genome. Genes Dev. 35, 749–770 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Bonnet, J. et al. PR-DUB preserves Polycomb repression by preventing excessive accumulation of H2Aub1, an antagonist of chromatin compaction. Genes Dev. 36, 1046–1061 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Beck, D. B. et al. Chromatin in the nuclear landscape. Cold Spring Harb. Symp. Quant. Biol. 75, 11–22 (2010).

    Article  CAS  PubMed  Google Scholar 

  11. Aloia, L., Di Stefano, B. & Di Croce, L. Polycomb complexes in stem cells and embryonic development. Development 140, 2525–2534 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. Chen, Z., Djekidel, M. N. & Zhang, Y. Distinct dynamics and functions of H2AK119ub1 and H3K27me3 in mouse preimplantation embryos. Nat. Genet. 53, 551–563 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Parreno, V., Martinez, A. M. & Cavalli, G. Mechanisms of Polycomb group protein function in cancer. Cell Res. 32, 231–253 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Gaytán de Ayala Alonso, A. et al. A genetic screen identifies novel polycomb group genes in Drosophila. Genetics 176, 2099–2108 (2007).

    Article  PubMed  Google Scholar 

  15. Harbour, J. W. et al. Frequent mutation of BAP1 in metastasizing uveal melanomas. Science 330, 1410–1413 (2010).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Pena-Llopis, S. et al. BAP1 loss defines a new class of renal cell carcinoma. Nat. Genet. 44, 751–579 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Dey, A. et al. Loss of the tumor suppressor BAP1 causes myeloid transformation. Science 337, 1541–1546 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  18. Carbone, M. et al. BAP1 and cancer. Nat. Rev. Cancer 13, 153–159 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Katoh, M. Functional and cancer genomics of ASXL family members. Br. J. Cancer 109, 299–306 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Wang, L., Birch, N. W., Zhao, Z., Nestler, C. M. & Shilatifard, A. Epigenetic targeted therapy of stabilized BAP1 in ASXL1 gain-of-function mutated leukemia. Nat. Cancer 2, 515–526 (2021).

    Article  PubMed  Google Scholar 

  21. Abdel-Wahab, O. et al. ASXL1 mutations promote myeloid transformation through loss of PRC2-mediated gene repression. Cancer Cell 22, 180–193 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Balasubramani, A. et al. Cancer-associated ASXL1 mutations may act as gain-of-function mutations of the ASXL1–BAP1 complex. Nat. Commun. 6, 7307 (2015).

    Article  ADS  CAS  PubMed  Google Scholar 

  23. Sahtoe, D. D., van Dijk, W. J., Ekkebus, R., Ovaa, H. & Sixma, T. K. BAP1/ASXL1 recruitment and activation for H2A deubiquitination. Nat. Commun. 7, 10292 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  24. De, I. et al. Structural basis for the activation of the deubiquitinase Calypso by the Polycomb protein ASX. Structure 27, 528–536 (2019).

    Article  CAS  PubMed  Google Scholar 

  25. Foglizzo, M. et al. A bidentate Polycomb repressive-deubiquitinase complex is required for efficient activity on nucleosomes. Nat. Commun. 9, 3932 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  26. Sahtoe, D. D. et al. Mechanism of UCH-L5 activation and inhibition by DEUBAD domains in RPN13 and INO80G. Mol. Cell 57, 887–900 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Vander Linden, R. T. et al. Structural basis for the activation and inhibition of the UCH37 deubiquitylase. Mol. Cell 57, 901–911 (2015).

    Article  Google Scholar 

  28. Tate, J. G. et al. COSMIC: the catalogue of somatic mutations in cancer. Nucleic Acids Res. 47, D941–D947 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. 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  PubMed Central  Google Scholar 

  30. Morgan, M. T. et al. Structural basis for histone H2B deubiquitination by the SAGA DUB module. Science 351, 725–728 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wang, H. et al. Structure of the transcription coactivator SAGA. Nature 577, 717–720 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  32. Davey, C. A., Sargent, D. F., Luger, K., Maeder, A. W. & Richmond, T. J. Solvent mediated interactions in the structure of the nucleosome core particle at 1.9 a resolution. J. Mol. Biol. 319, 1097–1113 (2002).

    Article  CAS  PubMed  Google Scholar 

  33. Lan, L. et al. Monoubiquitinated histone H2A destabilizes photolesion-containing nucleosomes with concomitant release of UV-damaged DNA-binding protein E3 ligase. J. Biol. Chem. 287, 12036–12049 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Nakata, S. et al. The dynamics of histone H2A ubiquitination in HeLa cells exposed to rapamycin, ethanol, hydroxyurea, ER stress, heat shock and DNA damage. Biochem. Biophys. Res. Commun. 472, 46–52 (2016).

    Article  CAS  PubMed  Google Scholar 

  35. Zlatanova, J. & Vanholde, K. Histone H1 and transcription—still an enigma. J. Cell Sci. 103, 889–895 (1992).

    Article  CAS  PubMed  Google Scholar 

  36. Jason, L. J. M., Finn, R. M., Lindsey, G. & Ausio, J. Histone H2A ubiquitination does not preclude histone H1 binding, but it facilitates its association with the nucleosome. J. Biol. Chem. 280, 4975–4982 (2005).

    Article  CAS  PubMed  Google Scholar 

  37. Zhou, B. R. et al. Distinct structures and dynamics of chromatosomes with different human linker histone isoforms. Mol. Cell 81, 166–182 (2021).

    Article  CAS  PubMed  Google Scholar 

  38. Zhu, P. et al. A histone H2A deubiquitinase complex coordinating histone acetylation and H1 dissociation in transcriptional regulation. Mol. Cell 27, 609–621 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lowary, P. T. & Widom, J. New DNA sequence rules for high affinity binding to histone octamer and sequence-directed nucleosome positioning. J. Mol. Biol. 276, 19–42 (1998).

    Article  CAS  PubMed  Google Scholar 

  40. Luger, K., Rechsteiner, T. J. & Richmond, T. J. Preparation of nucleosome core particle from recombinant histones. Methods Enzymol. 304, 3–19 (1999).

    Article  CAS  PubMed  Google Scholar 

  41. Fang, Q. et al. Human cytomegalovirus IE1 protein alters the higher-order chromatin structure by targeting the acidic patch of the nucleosome. eLife 5, e11911 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Dyer, P. N. et al. in Chromatin and Chromatin Remodeling Enzymes Part A (eds Allis, C.D. & Wu, C.) 23-44 (2004).

  43. Song, F. et al. Cryo-EM study of the chromatin fiber reveals a double helix twisted by tetranucleosomal units. Science 344, 376–380 (2014).

    Article  ADS  CAS  PubMed  Google Scholar 

  44. Kastner, B. et al. GraFix: sample preparation for single-particle electron cryomicroscopy. Nat. Methods 5, 53–55 (2008).

    Article  CAS  PubMed  Google Scholar 

  45. Schorb, M., Haberbosch, I., Hagen, W. J. H., Schwab, Y. & Mastronarde, D. N. Software tools for automated transmission electron microscopy. Nat. Methods 16, 471–477 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Wu, C., Huang, X., Cheng, J., Zhu, D. & Zhang, X. High-quality, high-throughput cryo-electron microscopy data collection via beam tilt and astigmatism-free beam-image shift. J. Struct. Biol. 208, 107396 (2019).

    Article  CAS  PubMed  Google Scholar 

  47. 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 

  48. Asarnow, D., Palovcak, E. & Cheng, Y. UCSF pyem v0.5. Zenodo https://doi.org/10.5281/zenodo.3576630 (2019).

  49. Zivanov, J. et al. New tools for automated high-resolution cryo-EM structure determination in RELION-3. eLife 7, e42166 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Sanchez-Garcia, R. et al. DeepEMhancer: a deep learning solution for cryo-EM volume post-processing. Commun. Biol. 4, 874 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Adams, P. D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D 66, 213–221 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Terwilliger, T. C., Ludtke, S. J., Read, R. J., Adams, P. D. & Afonine, P. V. Improvement of cryo-EM maps by density modification. Nat. Methods 17, 923–927 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Wang, R. Y. et al. Automated structure refinement of macromolecular assemblies from cryo-EM maps using Rosetta. eLife 5, e17219 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Chen, V. B. et al. MolProbity: all-atom structure validation for macromolecular crystallography. Acta Crystallogr. D 66, 12–21 (2010).

    Article  CAS  PubMed  Google Scholar 

  56. Pettersen, E. F. et al. UCSF Chimera—a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).

    Article  CAS  PubMed  Google Scholar 

  57. 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 

  58. Zhang, J. et al.Highly enriched BEND3 prevents the premature activation of bivalent genes during differentiation. Science 375, 1053–1058 (2022).

    Article  ADS  CAS  PubMed  Google Scholar 

  59. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Kim, D., Paggi, J. M., Park, C., Bennett, C. & Salzberg, S. L. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat. Biotechnol. 37, 907–915 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Pertea, M. et al. StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat. Biotechnol. 33, 290–295 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

  63. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank B. Zhu, X. Huang, L. Chen, X. Li and other staff members at the Center for Biological Imaging of the Institute of Biophysics (IBP), Chinese Academy of Sciences (CAS), for support in cryo-EM data collection; X. Yu at the Protein Science Core Facility of IBP for help with MALS analyses; Y. Gu for participation at an early stage of the study; and J. Chen for logistic support. The study was supported in part by grants from the Chinese Ministry of Science and Technology (2019YFA0508900), the Natural Science Foundation of China (31991162, 92153302 and 91853204), the K. C. Wong Education Foundation (GJTD-2020-06), the Youth Innovation Promotion Association (2018125) and the Strategic Priority Research Program (XDB37010100) of CAS.

Author information

Authors and Affiliations

Authors

Contributions

R.-M.X. & B.Z. designed and supervised the project. G.L. contributed experimental designs and advised on enzymatic activity assays. X.Z. advised on cryo-EM analysis. C.T. advised on the synthesis of ubiquitinated histones. W.G. performed protein expression and purification and cryo-EM sample preparation, analysed the structure and performed biochemical experiments. C.Y. prepared cryo-EM samples, collected data and solved and analysed the structures. J.L. analysed the cellular effects of BAP1 mutations. Z.Y. supervised the cryo-EM sample preparation and fluorescent polarization experiments. X.L. advised on enzymatic activity assays. Y.Z. performed bioinformatic analysis. C.-P.L. helped with structural model building. Y.L. contributed to the analysis of cellular effects of BAP1 mutations. W.G., C.Y. and R.-M.X. wrote the manuscript, and all authors read and edited the manuscript.

Corresponding authors

Correspondence to Bing Zhu or Rui-Ming Xu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Robert Klose and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Cryo-EM sample preparation.

a, Elution profile of size-exclusion column chromatography (SEC) of the BAP1–ASXL1(1–378) complex. Peak 1 was eluted close to the void volume of the column. b, Coomassie-stained SDS–PAGE analysis of fractions from SEC shown in (a). M, molecular weight marker; values are labelled on the left. Peak 2 fractions were collected for assembling complex with NCP. c, Western blot analyses of DUB activities of the wild-type and the C91S mutant BAP1 complexes with ASXL1(1–378). Same amount of the 187-bp DNA H2AK119ub1 chromatosome substrate was used in each reaction. From left to right, lane 1, no enzyme added; lanes 2–7 and lanes 8–13, successive twofold dilutions of the wild-type and C91S BAP1 enzyme complexes, respectively, detected with anti-H2A (top panel) and anti-His tag (bottom panel) antibodies. d, SEC-MALS analysis of BAP1 C91S–ASXL1(1–378) complex at the concentration of 2.2 mg/ml. Calculated molar mass = 1648.2 kDa, which is far below the 244.2 kDa calculated molecular mass of a dimer of the BAP1–ASXL1(1–378) complex. The data were analysed by the ASTRA software. e, GoldView-stained native PAGE shows the reconstituted NCP, NCP in a 600 mM NaCl buffer, at which point H1 was added and dialysed to zero salt to assemble the chromatosome. f, GoldView-stained native PAGE of fractions after GraFix. The chromatosome alone was used as a control. Fractions 12–14, highlighted in the red box, were used for cryo-EM sample preparation.

Source data

Extended Data Fig. 2 Structure determination of the BAP1–ASXL1(1–378)–H1–NCP(H2AK119ub1) complex.

a, A representative electron micrograph low-pass-filtered to 15 Å from 21,466 micrographs. b, Selected reference-free 2D class average. c, Workflow of cryo-EM data processing. d, The 3.0-Å global refinement map of the full complex (left), and the 3.9-Å local refinement BAP1–ASXL1(1–378)–ubiquitin subcomplex (right). For convenience of presentation, a composite map was constructed by combining indicated global and focused refinement maps. e, Angular distribution of the full complex particles in the final reconstructions. f, Gold-standard Fourier correlation of the BAP1–ASXL1(1–378)–H1–NCP(H2AK119ub1) complex.

Extended Data Fig. 3 EM density maps of various components or regions of the PR-DUB–substrate complex.

a, 601 nucleosomal DNA. b, The SHL 0 region of nucleosomal DNA. c, Human histone octamer. Two H3s are coloured in blue and light blue, H4s in pale cyan and pale green, H2As in yellow-orange and bright orange, and H2Bs in pink and light pink. d, The globular domain of linker histone H1.4. e, Human BAP1. f, The DEUBAD domain of ASXL1. g, The ubiquitin moiety of H2AK119ub1. h, Selected regions of core histones. i, BAP1 regions. j, The DEUBAD domain of ASXL1. k, The C-terminal region of the ubiquitin moiety of H2AK119ub1. Atomic models are shown in a stick representation, and the maps are contoured at 5σ~7σ.

Extended Data Fig. 4 Cryo-EM analysis of the BAP1–ASXL1(1–378)–NCP(H2AK119ub1) complex without H1.

a, A representative electron micrograph low-pass-filtered to 15 Å from 6,269 micrographs. b, Selected reference-free 2D class average. c, 2D class averages of two BAP1–ASXL1 complexes bound to NCP in higher contrast. The red arrows indicate the density for the bound BAP1–ASXL1 complexes. d, Workflow of cryo-EM data processing. e, Alignment of the EM densities for the BAP1–ASXL1(1–378)–NCP(H2AK119ub1) complexes with (blue) and without (grey) H1. f, 3D classification map of two BAP1–ASXL1(1–378) complexes bound to the nucleosome.

Extended Data Fig. 5 BAP1 residues in distinct regions involved in H2AK119 deubiquitination and nucleosome binding.

a, Sequence alignment of the CTE region of BAP1 orthologues. The RRRQ (aa 699–702) CTE finger motif is boxed in dashed lines. b, Alignment of the RRSRR motif (aa 56–60) in the β2–α2 loop of BAP1. c, Alignment of the sequences encompassing the α8–β6 loop of BAP1, Caly and UCH-L5. Above the sequence, BAP1 secondary structure elements are depicted, and period signs denote whole 10th residues of BAP1. df, Quantification of catalytic activities of western blot measurements in Figs. 2d, g and h, respectively. The values for indicated wild-type and BAP1 mutant complexes measured at 5 and 40 min in Fig. 2d,h are shown with clear and grey-shaded bars, respectively. The wild-type values (at 40 min) were set to unity, and all values were derived from three replicates, and represented as mean ± s.e.m. P values, displayed at the top of the bar charts, denote the results of an unpaired two-tailed t-test. g, Fluorescence polarization measurements of binding affinities of the BAP1–ASXL1 complexes carrying indicated BAP1 (56–60 mut) or ASXL1 (T262W, 1–330) mutations to the NCP(H2AK119ub1) assembled with 167-bp 601 DNA containing a 5’-FAM fluorophore. The data were analysed in the same way as in Fig. 2e (n = 3 biologically independent experiments; mean ± s.e.m.). h, Western blot analysis of free histone H2AK119 deubiquitination by wild-type and indicated BAP1 mutants in complex with ASXL1(1–378). A ‘−’ sign indicates a mock reaction without enzyme added. Reactions were monitored at the indicated time points, and detected with anti-His tag (top) or anti-H2A (bottom) antibodies.

Source data

Extended Data Fig. 6 Gene set enrichment analysis.

a,b, Upregulated (a) and downregulated (b) genes in Bap1 KO ES cells versus wild-type ES cells from a previous study7 with respect to the global transcriptional changes observed in our Bap1 KO + EV ES cells versus wild-type ES cells. c,d, Upregulated (c) and downregulated (d) genes analysed in a previous study 8 relative to the global transcriptional changes we observed.

Extended Data Fig. 7 Gene expression analysis of Bap1 mutants.

a, Percentage of rescued BAP1-regulated genes (differentially expressed genes between Bap1 KO + EV ES cells and wild-type ES cells) in the indicated wild-type and mutant BAP1 rescue cells. b, Expression correlation among the 14 RNA-seq samples of mouse ES cells on BAP1-regulated genes (differentially expressed genes between Bap1 KO + EV and wild-type R1). There are two replicates for each cell line. Pearson correlation coefficients are indicated.

Extended Data Fig. 8 Cellular DUB activities and changes in gene expression for BAP1 CTE cancer-associated mutations.

a, Western blot detection of the levels of ubH2A and BAP1 in Bap1 KO cells rescued with the indicated BAP1 mutant plasmids. Quantification of the western blot results is represented in a bar chart of mean ± s.e.m. (n = 4 biologically independent experiments). P values, displayed at the top of the bar chart, denote the results of an unpaired two-tailed t-test. b, Heat maps of transcriptional levels of BAP1-regulated genes in wild-type and indicated rescue cell lines. c, Percentage of rescued BAP1-regulated genes (differentially expressed genes between Bap1 KO and wild-type ES cells) in the indicated mutant BAP1 rescue cells. d, Expression correlation among the 10 RNA-seq samples.

Source data

Extended Data Fig. 9 Quantification of cellular DUB activities and RNA-seq analysis for non-CTE BAP1 cancer-associated mutations.

a, Quantification of western blot detection of H2AK119ub and BAP1, as shown in Fig. 3h, in Bap1 KO cells transfected with an empty vector (EV), or plasmids re-expressing wild-type or the indicated BAP1 mutants. Bar charts represent plots of mean ± s.e.m. (n = 4 biologically independent experiments). P values, displayed at the top of the bar chart, denote the results of an unpaired two-tailed t-test. b, Heat maps of transcriptional levels of BAP1-regulated genes in wild-type and indicated rescue cell lines. There are two replicates for each cell line. c, Percentage of rescued BAP1-regulated genes (differentially expressed genes between Bap1 KO and wild-type ES cells) in each indicated mutant BAP1 rescue cells. d, Expression correlation among the 10 RNA-seq samples.

Source data

Extended Data Fig. 10 Structure and function of ASXL1 domains and the effect of H1 on H2AK119 deubiquitination.

a, Domain structures of ASXL1–ASXL3 and alignment of their N-terminal sequences encompassing HARE-HTH and DEUBAD domains. b, Left, schematic representation of ASXL1 constructs used in the deubiquitinase assay. Wavy lines represent a 2×(GGGGS) linker. Middle, western blot analysis of nucleosomal H2AK119 deubiquitination of BAP1 complexes with the indicated ASXL1 fragments. H3 lanes are sample-processing controls. Right, quantification of catalytic activities based on western blot measurements (n = 3 biologically independent experiments, mean ± s.e.m.). P values, displayed at the top of the bar chart, denote the results of an unpaired two-tailed t-test. No adjustment was made for multiple comparisons. c, A BAP1–ASXL1(1–378) complex modelled to the proximal DNA side (left) of the chromatosome based on the observed binding mode in the distal DNA side (right) sterically clashes with H1 (see inset).

Source data

Extended Data Table 1 Cryo-EM data collection, refinement and validation statistics

Supplementary information

Supplementary Information

This file contains the sequences for mutagenesis primers and nucleosomal DNA (Supplementary Table 1) and the uncropped blots and FACS gating strategy (Supplementary Figs. 1 and 2).

Reporting Summary

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

Ge, W., Yu, C., Li, J. et al. Basis of the H2AK119 specificity of the Polycomb repressive deubiquitinase. Nature 616, 176–182 (2023). https://doi.org/10.1038/s41586-023-05841-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-023-05841-y

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