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:

mSWI/SNF promotes Polycomb repression both directly and through genome-wide redistribution

Abstract

The mammalian SWI/SNF complex, or BAF complex, has a conserved and direct role in antagonizing Polycomb-mediated repression. Yet, BAF also promotes repression by Polycomb in stem cells and cancer. How BAF both antagonizes and promotes Polycomb-mediated repression remains unknown. Here, we utilize targeted protein degradation to dissect the BAF–Polycomb axis in mouse embryonic stem cells on short timescales. We report that rapid BAF depletion redistributes Polycomb repressive complexes PRC1 and PRC2 from highly occupied domains, like Hox clusters, to weakly occupied sites normally opposed by BAF. Polycomb redistribution from highly repressed domains results in their decompaction, gain of active epigenomic features and transcriptional derepression. Surprisingly, through dose-dependent degradation of PRC1 and PRC2, we identify a conventional role for BAF in Polycomb-mediated repression, in addition to global Polycomb redistribution. These findings provide new mechanistic insight into the highly dynamic state of the Polycomb–Trithorax axis.

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: Brg1 degradation results in the derepression of many highly Polycomb-bound genes.
Fig. 2: PRC1 and PRC2 are quickly redistributed following Brg1 degradation.
Fig. 3: Brg1 degradation and Polycomb loss result in decompaction of HoxA and HoxD regions.
Fig. 4: Genome-wide Polycomb redistribution changes chromatin state from repressed to active.
Fig. 5: BAF promotes repression directly and by genome-wide Polycomb redistribution.
Fig. 6: Increased PRC1 dosage inhibits Brg1 degron-mediated derepression.
Fig. 7: Model and summary of findings.

Similar content being viewed by others

Data availability

All sequencing data have been deposited in the GEO (accession no. GSE145016). Source data are provided with this paper.

References

  1. Flavahan, W. A., Gaskell, E. & Bernstein, B. E. Epigenetic plasticity and the hallmarks of cancer. Science 357, eaal2380 (2017).

    Article  PubMed  PubMed Central  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. Kingston, R. E. & Tamkun, J. W. Transcriptional regulation by trithorax-group proteins. Cold Spring Harb. Perspect. Biol. 6, a019349 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Kennison, J. A. & Tamkun, J. W. Dosage-dependent modifiers of Polycomb and antennapedia mutations in Drosophila. Proc. Natl Acad. Sci. USA 85, 8136–8140 (1988).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Tamkun, J. W. et al. brahma: a regulator of Drosophila homeotic genes structurally related to the yeast transcriptional activator SNF2/SWI2. Cell 68, 561–572 (1992).

    Article  CAS  PubMed  Google Scholar 

  6. Yu, B. D., Hess, J. L., Horning, S. E., Brown, G. A. & Korsmeyer, S. J. Altered Hox expression and segmental identity in Mll-mutant mice. Nature 378, 505–508 (1995).

    Article  CAS  PubMed  Google Scholar 

  7. Kadoch, C. & Crabtree, G. R. Reversible disruption of mSWI/SNF (BAF) complexes by the SS18–SSX oncogenic fusion in synovial sarcoma. Cell 153, 71–85 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Shain, A. H. & Pollack, J. R. The spectrum of SWI/SNF mutations, ubiquitous in human cancers. PLoS ONE 8, e55119 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Ronan, J. L., Wu, W. & Crabtree, G. R. From neural development to cognition: unexpected roles for chromatin. Nat. Rev. Genet. 14, 347–359 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Clapier, C. R., Iwasa, J., Cairns, B. R. & Peterson, C. L. Mechanisms of action and regulation of ATP-dependent chromatin-remodelling complexes. Nat. Rev. Mol. Cell Biol. 18, 407–422 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kadoch, C. et al. Dynamics of BAF–Polycomb complex opposition on heterochromatin in normal and oncogenic states. Nat. Genet. 49, 213–222 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Stanton, B. Z. et al. Smarca4 ATPase mutations disrupt direct eviction of PRC1 from chromatin. Nat. Genet. 49, 282–288 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Braun, S. M. G. et al. Rapid and reversible epigenome editing by endogenous chromatin regulators. Nat. Commun. 8, 560 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Valencia, A. M. & Kadoch, C. Chromatin regulatory mechanisms and therapeutic opportunities in cancer. Nat. Cell Biol. 21, 152–161 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. McBride, M. J. et al. The SS18–SSX fusion oncoprotein hijacks BAF complex targeting and function to drive synovial sarcoma. Cancer Cell 33, 1128–1141 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Johann, P. D. et al. Atypical teratoid/rhabdoid tumors are comprised of three epigenetic subgroups with distinct enhancer landscapes. Cancer Cell 29, 379–393 (2016).

    Article  CAS  PubMed  Google Scholar 

  17. Alexander, J. M. et al. Brg1 modulates enhancer activation in mesoderm lineage commitment. Development 142, 1418–1430 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Ho, L. et al. esBAF facilitates pluripotency by conditioning the genome for LIF/STAT3 signalling and by regulating Polycomb function. Nat. Cell Biol. 13, 903–913 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Nakayama, R. T. et al. SMARCB1 is required for widespread BAF complex-mediated activation of enhancers and bivalent promoters. Nat. Genet. 49, 1613–1623 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Masliah-Planchon, J., Bièche, I., Guinebretière, J.-M., Bourdeaut, F. & Delattre, O. SWI/SNF chromatin remodeling and human malignancies. Annu. Rev. Pathol. 10, 145–171 (2015).

    Article  CAS  PubMed  Google Scholar 

  21. Nishimura, K., Fukagawa, T., Takisawa, H., Kakimoto, T. & Kanemaki, M. An auxin-based degron system for the rapid depletion of proteins in nonplant cells. Nat. Methods 6, 917–922 (2009).

    Article  CAS  PubMed  Google Scholar 

  22. Morawska, M. & Ulrich, H. D. An expanded tool kit for the auxin-inducible degron system in budding yeast. Yeast 30, 341–351 (2013).

    Article  CAS  PubMed  Google Scholar 

  23. Nora, E. P. et al. Targeted degradation of CTCF decouples local insulation of chromosome domains from genomic compartmentalization. Cell 169, 930–944 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Joshi, O. et al. Dynamic reorganization of extremely long-range promoter–promoter interactions between two states of pluripotency. Cell Stem Cell 17, 748–757 (2015).

    Article  CAS  PubMed  Google Scholar 

  25. Dykhuizen, E. C., Carmody, L. C., Tolliday, N., Crabtree, G. R. & Palmer, M. A. Screening for inhibitors of an essential chromatin remodeler in mouse embryonic stem cells by monitoring transcriptional regulation. J. Biomol. Screen. 17, 1221–1230 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Boyer, L. A. et al. Polycomb complexes repress developmental regulators in murine embryonic stem cells. Nature 441, 349–353 (2006).

    Article  CAS  PubMed  Google Scholar 

  27. Ernst, J. & Kellis, M. ChromHMM: automating chromatin-state discovery and characterization. Nat. Methods 9, 215–216 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Mateo, L. J., Sinnott-Armstrong, N. & Boettiger, A. N. Tracing DNA paths and RNA profiles in cultured cells and tissues with ORCA. Nat. Protoc. 16, 1647–1713 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Eskeland, R. et al. Ring1B compacts chromatin structure and represses gene expression independent of histone ubiquitination. Mol. Cell 38, 452–464 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Francis, N. J., Kingston, R. E. & Woodcock, C. L. Chromatin compaction by a Polycomb group protein complex. Science 306, 1574–1577 (2004).

    Article  CAS  PubMed  Google Scholar 

  31. Bintu, B. et al. Super-resolution chromatin tracing reveals domains and cooperative interactions in single cells. Science 362, eaau1783 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Mateo, L. J. et al. Visualizing DNA folding and RNA in embryos at single-cell resolution. Nature 568, 49–54 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Bonev, B. et al. Multiscale 3D genome rewiring during mouse neural development. Cell 171, 557–572 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Piunti, A. & Shilatifard, A. Epigenetic balance of gene expression by Polycomb and COMPASS families. Science 352, aad9780 (2016).

    Article  PubMed  Google Scholar 

  35. Dallas, P. B. et al. p300/CREB binding protein-related protein p270 is a component of mammalian SWI/SNF complexes. Mol. Cell. Biol. 18, 3596–3603 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Tie, F. et al. CBP-mediated acetylation of histone H3 lysine 27 antagonizes Drosophila Polycomb silencing. Development 136, 3131–3141 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Hainer, S. J. et al. Suppression of pervasive noncoding transcription in embryonic stem cells by esBAF. Genes Dev. 29, 362–378 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Wang, X. et al. Molecular analysis of PRC2 recruitment to DNA in chromatin and its inhibition by RNA. Nat. Struct. Mol. Biol. 24, 1028–1038 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Bensaude, O. Inhibiting eukaryotic transcription: which compound to choose? How to evaluate its activity? Transcription 2, 103–108 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Riising, E. M. et al. Gene silencing triggers Polycomb repressive complex 2 recruitment to CpG islands genome wide. Mol. Cell 55, 347–360 (2014).

    Article  CAS  PubMed  Google Scholar 

  41. Francis, N. J., Saurin, A. J., Shao, Z. & Kingston, R. E. Reconstitution of a functional core Polycomb repressive complex. Mol. Cell 8, 545–556 (2001).

    Article  CAS  PubMed  Google Scholar 

  42. Shao, Z. et al. Stabilization of chromatin structure by PRC1, a Polycomb complex. Cell 98, 37–46 (1999).

    Article  CAS  PubMed  Google Scholar 

  43. Endoh, M. et al. Polycomb group proteins Ring1A/B are functionally linked to the core transcriptional regulatory circuitry to maintain ES cell identity. Development 135, 1513–1524 (2008).

    Article  CAS  PubMed  Google Scholar 

  44. Miller, E. L. et al. TOP2 synergizes with BAF chromatin remodeling for both resolution and formation of facultative heterochromatin. Nat. Struct. Mol. Biol. 24, 344–352 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. King, H. W., Fursova, N. A., Blackledge, N. P. & Klose, R. J. Polycomb repressive complex 1 shapes the nucleosome landscape but not accessibility at target genes. Genome Res. 28, 1494–1507 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Hodges, H. C. et al. Dominant-negative SMARCA4 mutants alter the accessibility landscape of tissue-unrestricted enhancers. Nat. Struct. Mol. Biol. 25, 61–72 (2018).

    Article  CAS  PubMed  Google Scholar 

  47. Nabet, B. et al. The dTAG system for immediate and target-specific protein degradation. Nat. Chem. Biol. 14, 431–441 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Blackledge, N. P. et al. PRC1 catalytic activity is central to Polycomb system function. Mol. Cell 77, 857–874 (2019).

    Article  PubMed  Google Scholar 

  49. Tamburri, S. et al. Histone H2AK119 mono-ubiquitination is essential for Polycomb-mediated transcriptional repression. Mol. Cell 77, 840–856 (2019).

    Article  PubMed  Google Scholar 

  50. Fursova, N. A. et al. Synergy between variant PRC1 complexes defines Polycomb-mediated gene repression. Mol. Cell 74, 1020–1036 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Kumar, B. & Elsasser, S. J. Quantitative multiplexed ChIP reveals global alterations that shape promoter bivalency in ground state embryonic stem cells. Cell Rep. 28, 3274–3284 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. van Mierlo, G. et al. Integrative proteomic profiling reveals PRC2-dependent epigenetic crosstalk maintains ground-state pluripotency. Cell Stem Cell 24, 123–137 (2019).

    Article  PubMed  Google Scholar 

  53. Bracken, A. P., Brien, G. L. & Verrijzer, C. P. Dangerous liaisons: interplay between SWI/SNF, NuRD and Polycomb in chromatin regulation and cancer. Genes Dev. 33, 936–959 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Yildirim, O. et al. Mbd3/NURD complex regulates expression of 5-hydroxymethylcytosine marked genes in embryonic stem cells. Cell 147, 1498–1510 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Conrad, R. J. et al. The short isoform of BRD4 promotes HIV-1 latency by engaging repressive SWI/SNF chromatin-remodeling complexes. Mol. Cell 67, 1001–1012 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Karczewski, K. J. et al. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature 581, 434–443 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. He, S. et al. Structure of nucleosome-bound human BAF complex. Science 367, 875–881 (2020).

    Article  CAS  PubMed  Google Scholar 

  58. Wollmann, P. et al. Structure and mechanism of the Swi2/Snf2 remodeller Mot1 in complex with its substrate TBP. Nature 475, 403–407 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Fonseca, J. P. et al. In vivo Polycomb kinetics and mitotic chromatin binding distinguish stem cells from differentiated cells. Genes Dev. 26, 857–871 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Kadoch, C. & Crabtree, G. R. Mammalian SWI/SNF chromatin remodeling complexes and cancer: mechanistic insights gained from human genomics. Sci. Adv. 1, e1500447 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Philippidou, P. & Dasen, J. S. Hox genes: choreographers in neural development, architects of circuit organization. Neuron 80, 12–34 (2013).

    Article  CAS  PubMed  Google Scholar 

  62. Kidder, B. L., Palmer, S. & Knott, J. G. SWI/SNF-Brg1 regulates self-renewal and occupies core pluripotency-related genes in embryonic stem cells. Stem Cells 27, 317–328 (2009).

    Article  CAS  PubMed  Google Scholar 

  63. Reilly, M. B., Cros, C., Varol, E., Yemini, E. & Hobert, O. Unique homeobox codes delineate all the neuron classes of C. elegans. Nature 584, 595–601 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Ying, Q. L. et al. The ground state of embryonic stem cell self-renewal. Nature 453, 519–523 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Martin, M. CUTADAPT removes adapter sequences from high-throughput sequencing reads. EMBnet J. https://doi.org/10.14806/ej.17.1.200 (2011).

  66. Bray, N. L., Pimentel, H., Melsted, P. & Pachter, L. Near-optimal probabilistic RNA-seq quantification. Nat. Biotechnol. 34, 525–527 (2016).

    Article  CAS  PubMed  Google Scholar 

  67. Soneson, C., Love, M. I. & Robinson, M. D. Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. F1000Res. 4, 1521 (2015).

    Article  PubMed  Google Scholar 

  68. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Stephens, M. False discovery rates: a new deal. Biostatistics 18, 275–294 (2017).

    PubMed  Google Scholar 

  70. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Ramirez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Robinson, J. T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24–26 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Raudvere, U. et al. g:Profiler: a web server for functional enrichment analysis and conversions of gene lists (2019 update). Nucleic Acids Res. 47, W191–W198 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Amemiya, H. M., Kundaje, A. & Boyle, A. P. The ENCODE blacklist: identification of problematic regions of the genome. Sci. Rep. 9, 9354 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  76. Wickham, H. ggplot2: Elegant Graphics for Data Analysis 2nd edn (Springer, 2016).

  77. Durand, N. C. et al. Juicer provides a one-click system for analyzing loop-resolution Hi-C experiments. Cell Syst. 3, 95–98 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Robinson, J. T. et al. Juicebox.js provides a cloud-based visualization system for Hi-C data. Cell Syst. 6, 256–258 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Gatchalian, J. et al. A non-canonical BRD9-containing BAF chromatin remodeling complex regulates naive pluripotency in mouse embryonic stem cells. Nat. Commun. 9, 5139 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  80. de Dieuleveult, M. Genome-wide nucleosome specificity and function of chromatin remodellers in ES cells. Nature 530, 113–116 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of the Crabtree laboratory for insightful comments and discussions over the course of the study. We are grateful to A. Krokhotin, S. Ramachandran (CU Anschutz) and V. Ramani (UCSF) for providing critical comments on the manuscript and for helpful discussions. This study was supported by NIH grants R01CA163915 (G.R.C.), R37NS046789 (G.R.C.) and DP2GM132935 (A.N.B.); the Howard Hughes Medical Institute (G.R.C.); a Swiss National Science Foundation (SNSF) postdoctoral fellowship (S.M.G.B.); the Walter V. and Idun Berry Postdoctoral fellowship program (C.M.W. and A.H.); and the Sir James Black postdoctoral fellowship (C.M.W. and S.M.G.B.).

Author information

Authors and Affiliations

Authors

Contributions

C.M.W. conceived the project and carried out experiments and analysis. A.H. performed ORCA experiments and analysis. S.M.G.B., J.G.K. and B.Z.S. performed experiments and analysis. G.R.C. and A.N.B. designed experiments, provided valuable insight and supervised the project. C.M.W. wrote the manuscript with assistance from all authors.

Corresponding author

Correspondence to Gerald R. Crabtree.

Ethics declarations

Competing interests

G.R.C. is founder and stockholder of Foghorn Therapeutics. The other authors declare no competing interests.

Additional information

Peer review information Nature Structural & Molecular Biology thanks Robert Illingworth and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available. Beth Moorefield was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Extended characterization of Brg1-AID degron, related to Fig. 1.

(a, Genotyping PCR for three homozygous knock-in clones (AID*20 was used for most experiments in the manuscript). b, Western blot of whole-cell extracts from knock-in lines showing 8.8kDa shift in migration from c-terminal tag (G4S)3-AID*-G4S-3×FLAG. Line 20 and 26 have homozygous insertions in frame that were also confirmed by Sanger sequencing. c, Representative western blot and quantitation of Brg1 abundance in parent line compared to Brg1-AID* ± osTIR1 induction (-auxin). Error bars represent mean ± standard deviation from five biological replicates. d, Comparison of Brg1 depletion kinetics between AID and Cre-lox genetic deletion by western blot and changes to colony morphology that occur at the 24-hour time-point. Cells were only grown on the same dish for 72 h to illustrate timing for Brg1fl/fl colony morphology changes. Experiment was repeated at least 3 times with similar results. Scale bar = 50 µm. e, Alkaline phosphatase staining of Brg1 AID* 20 line on gelatin coated dishes, showing that changes to colony morphology occur ~24 h. Top scale bar = 200 µm, bottom scale bar = 50 µm. Experiment was repeated at least 3 times with similar results.

Source data

Extended Data Fig. 2 Extended summary of RNA-seq results, related to Fig. 1.

a, Volcano plots depicting gene expression changes at 0.5, 1, 2, 4, and 8 hours of Brg1 degradation. Differentially expressed genes are colored red (FDR-corrected P < 0.05) from four biological replicates for each time-point. b, Pbrm1 degradation with auxin, does not derepress Hox genes, revealing that pBAF is not required for their repression and controls for auxin, Tir1 expression, and proteasomal degradation in the Brg1 degron. Error bars are mean ± standard error for n = 3 replicates. c, Gene set enrichment analysis for differentially downregulated genes at 4 and 8-hour timepoints. d, Overlap of differentially expressed genes in 72 h Brg1 fl/fl from12 and 8 h Brg1 AID calculated by Fisher’s exact test (p-value = 1.1e-46). e, Strong lentiviral Bmi1 overexpression doesn’t derepress Hox genes or other genes derepressed by Brg1 degron. Error bars depict standard error of the mean from three biological replicates.

Source data

Extended Data Fig. 3 Brg1 degradation results in quick redistribution of PRC1&2 but doesn’t affect net dosage, related to Fig. 2.

a, Principal component analysis of Ring1b and Suz12 ChIP-seq (n = 4 replicates) b, Browser snapshot of the difference in normalized counts (8 h – 0 h) for Ring1b and Suz12 at Hox A, B, C, and D depicting net loss across all four clusters. c, Average metagene plots of normalized counts (to read-depth) at sites that were significantly increased or decreased by DESeq2 analysis (FDR-corrected P < 0.1) and normalized to peak intensity, providing additional confirmation that normalization is correct. d, ChIP qPCR timecourse at 4, 8, and 24 h Brg1 degradation for Ring1b at sites that were significantly decreased by ChIP-seq. Error bars depict standard error of the mean from four biological replicates. (* = p < 0.05, ** = p < 0.005, *** = p < 0.0005; two-sided t test with Holm-Sidak multiple comparison correction). e, (left) Representative western blot showing Brg1 degradation efficiency and that H2AK119ub and H3K27me3 marks don’t change at 8 h time-point (ChIP-seq time-point for the complexes that deposit these marks). (right) Representative western showing Brg1 degradation efficiency over a longer timecourse and that Suz12, Ring1b, and H3K27me3 marks don’t noticeably change even at 48 h. Experiment was repeated at least 2 times with similar results (f) Correlation of all Ring1b and Suz12 peaks that are ± 2kb from gene transcription start sites and expression changes at 8 h Brg1 degradation. Correlation and p-values were obtained from Pearson’s product moment correlation. g, chromatin state enrichment for all, increased, and decreased peak changes for Ring1b and Suz12. h, ChIP qPCR and qRT-PCR validation of genes that gain Ring1b and Suz12 and have reduced gene expression following 8 h Brg1 degradation. Error bars depict standard error of the mean from five biological replicates.

Source data

Extended Data Fig. 4 Comparison of ORCA to Hi-C, related to Fig. 3.

a, Average contact frequency as measured by high resolution Hi-C from serum grown cells33. b, Average contact frequency as measured by ORCA (200-nm threshold). c, Pearson’s correlation and Pearson’s r were computed using all unique pairwise combinations of X,Y barcodes measured. d, Histograms depicting the difference in median distance for barcode pair combinations at HoxA. e, Histograms depicting the difference in median distance for barcode pair combinations at HoxD.

Extended Data Fig. 5 Extended analysis related to Fig. 4.

a, Heatmap depicting the negative correlation between H3K4me3 and Ring1b and Suz12 as the difference in normalized counts (8 h – 0 h) at peaks (8,220) that intersect H3K4me3, Ring1b, and Suz12 at merged peak center, sorted by decreasing H3K4me3 signal. b, Heatmap depicting the negative correlation between H3K27ac and PRC1&2 as the difference in normalized counts (8 h – 0 h) at peaks (4,140) that intersect H3K27ac, Ring1b, and Suz12 at merged peak center, sorted by decreasing H3K27ac signal. c,d, Scatterplots showing negative correlation between ATAC-seq changes (KO/control)44 and Ring1b (8 h/0 h) or Suz12 (8 h/0 h).

Extended Data Fig. 6 Transcription is not required for polycomb redistribution.

a, Experimental design to test whether polycomb redistribution requires transcription. b, qRT-PCR depicting fold change by ΔΔCt method with indicated treatments from n = 3 biological replicates. c,d, Scatterplot showing that Ring1b and Suz12 changes following Brg1 degradation are highly correlated despite global inhibition of transcription for all peaks c, and peaks that were significantly changed by Brg1 degradation alone (d) (FDR < 0.1). e, Scatterplot of changes to Ring1b and Suz12 upon treatment with auxin (auxin / control) or triptolide (triptolide / control) showing distinct effects from the two treatments. Correlation and p-values were obtained from Pearson’s product moment correlation for c-e, from n = 4 biological replicates at the 8 h timepoint.

Extended Data Fig. 7 PRC1 does not restrict BAF binding despite extensive overlap.

a, Heatmaps depicting Ring1b, Suz12, BAF155, and Brg1 ChIPseq signal from79 at peaks (8,228) that overlap Ring1b and Suz12, sorted by decreasing Ring1b signal (left) or Brg1 peaks (97,987) sorted by decreasing Brg1 signal (right). b, High overlap between Brg1 and BAF155 despite different number of peaks called due to antibody quality and read depth. c, BAF155 ChIP-seq in Ring1bfl/fl x Ring1a-/- x ActinCreER ESCs treated with Tamoxifen or EtOH for 72 h. MA plot (TAM/EtOH) highlights large number of unchanged peaks (>95% in blue) (n = 2). d, Analysis of the chromatin features that characterize BAF155 binding sites reveal that Ring1a/b double KO does not alter BAF complex binding, as the complex remains mainly bound to enhancers and bivalent promoters in both control and mutant ESCs. e, Representative genome tracks showing similar BAF155 levels across the Lefty1/2 locus in Tamoxifen and EtOH treated cells. f, Metagene plot and heatmaps showing that BAF155 ChIPseq signal is minimally affected by Ring1b deletion (g) Characterization of BAF155 peaks in EtOH and Tamoxifen conditions. Of the 49,000 detected BAF155 peaks in this dataset, less than 5% were significantly changed upon Ring1a/b double KO in ESCs and the small number of gained and decreased sites was similar. Despite 56% of Ring1b peaks overlapping with a BAF peak, only 1% of differentially bound peaks are within a PRC1 domain (bottom).

Extended Data Fig. 8 Characterization of EED/Ring1b degron and extended analysis related to Fig. 5.

a, Schematic depicting EED and Ring1b dTAG targeted protein degradation strategy in mESCs, where EED is tagged at the C-terminus and Ring1b is tagged at the N-terminus with FKBPF36V to enable targeted degradation with dTAG13 PROTAC. b, Representative western blot and quantitation of EED and Ring1b abundance in parent line compared to lines tagged with FKBPF36V. Error bars depict mean ± standard error of the mean from three tagged EED lines and four tagged Ring1b lines. c, Genotyping PCR and western from whole cell extract showing 14.6kDa shift in migration from C-terminus (G4S)3-FKBPF36V-G4S-V5 tag on three different EED clones used for experiments. d, Genotyping PCR and western from whole cell extract with 14.2kDa shift in migration from N-terminus FKBPF36V-G4S-HA-(G4S)3. Experiment was repeated at least 2 times with similar results (e) Brg1 ChIPseq showing localization to the TSS of differentially expressed genes, ChIP from79. f, Brg1 MNase ChIPseq showing localization to the TSS of differentially expressed genes, ChIP from80. g, Brg1 MNase ChIPseq showing higher occupancy at PRC bound genes repressed by Brg1 (independent of polycomb). Compare to Fig. 5f. h, Brg1 represses accessibility at PRC bound genes derepressed by Brg1 degron and Brg1/PRC degron. Accessibility change (Brg1 KO / control) by overlap (Fig. 6f) for peaks ± 2kb from gene promoter, ATAC data from44 with two-sided t-test p-value above. (i,top) Higher and broader polycomb domains over genes derepressed by both Brg1 and PRC degron than genes derepressed by Brg1 degron alone, with Suz12 occupancy shown here. (i, bottom) Broad domains over genes derepressed by both Brg1 and PRC degron show substantially more redistribution in the Brg1 degron. Compare to Ring1b signal in Fig. 6. (j) Expression difference (Brg1 – PRC Log2 Fold Change) for genes derepressed by Brg1 and PRC degron, over dosage titration (Most dosage sensitive genes are shown in Fig. 6g), from n = 4 biological replicates. Boxplot bounds depict quartile 1, median, and quartile 3 with whiskers at 1.5× interquartile range.

Extended Data Fig. 9 Histone modifications deposited by PRC1 and PRC2 are higher in 2i growth conditions and results in weaker derepression, related to Figs. 2 and 6.

a, Western blot and relative quantification of H2AK119ub and H3K27me3 normalized to H3 in cells grown in Serum/LIF or 2i growth conditions from n = 3 biological replicates. b, Hox gene derepression measured by qRT-PCR for cells grown in serum, 2i, and the ratio of serum/2i from n = 2 biological replicates.

Source data

Supplementary information

Source data

Source Data Fig. 1

Blot supporting Fig. 1.

Source Data Fig. 5

Blot supporting Fig. 5.

Source Data Fig. 6

Blot supporting Fig. 6.

Source Data Extended Data Fig. 1

Blot supporting Extended Fig. 1.

Source Data Extended Data Fig. 2

Blot supporting Extended Fig. 2.

Source Data Extended Data Fig. 3

Blot supporting Extended Fig. 3.

Source Data Extended Data Fig. 9

Blot supporting Extended Fig. 9.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Weber, C.M., Hafner, A., Kirkland, J.G. et al. mSWI/SNF promotes Polycomb repression both directly and through genome-wide redistribution. Nat Struct Mol Biol 28, 501–511 (2021). https://doi.org/10.1038/s41594-021-00604-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-021-00604-7

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