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:

Dynamics of BAF–Polycomb complex opposition on heterochromatin in normal and oncogenic states

Abstract

The opposition between Polycomb repressive complexes (PRCs) and BAF (mSWI/SNF) complexes has a critical role in both development and disease. Mutations in the genes encoding BAF subunits contribute to more than 20% of human malignancies, yet the underlying mechanisms remain unclear, owing largely to a lack of assays to assess BAF function in living cells. To address this, we have developed a widely applicable recruitment assay system through which we find that BAF opposes PRC by rapid, ATP-dependent eviction, leading to the formation of accessible chromatin. The reversal of this process results in reassembly of facultative heterochromatin. Surprisingly, BAF-mediated PRC eviction occurs in the absence of RNA polymerase II (Pol II) occupancy, transcription, and replication. Further, we find that tumor-suppressor and oncogenic mutant BAF complexes have different effects on PRC eviction. The results of these studies define a mechanistic sequence underlying the resolution and formation of facultative heterochromatin, and they demonstrate that BAF opposes PRC on a minute-by-minute basis to provide epigenetic plasticity.

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

Access options

Buy this article

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

Figure 1: Design and development of a rapidly inducible system to recruit BAF complexes to heterochromatin in living cells.
Figure 2: BAF complexes displace PRC repression upon recruitment.
Figure 3: Rapid removal of BAF complexes by competitive inhibition of rapamycin triggers reformation of repressed heterochromatin.
Figure 4: BAF complex-mediated eviction of Polycomb is ATP dependent.
Figure 5: SMARCB1-depleted (MRT-like) BAF complexes fail to oppose PRC repression.
Figure 6: SS18-SSX-containing (synovial sarcoma–like) BAF complexes exhibit enhanced occupancy and Polycomb eviction.
Figure 7: Model for rapid BAF–Polycomb opposition in normal and oncogenic settings.

Similar content being viewed by others

References

  1. Kennison, J.A. & Tamkun, J.W. Trans. -regulation of homeotic genes in Drosophila. New Biol. 4, 91–96 (1992).

    CAS  PubMed  Google Scholar 

  2. Simon, J.A. & Kingston, R.E. Occupying chromatin: Polycomb mechanisms for getting to genomic targets, stopping transcriptional traffic, and staying put. Mol. Cell 49, 808–824 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Morgan, M.A. & Shilatifard, A. Chromatin signatures of cancer. Genes Dev. 29, 238–249 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  5. Margueron, R. & Reinberg, D. The Polycomb complex PRC2 and its mark in life. Nature 469, 343–349 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kadoch, C. et al. Proteomic and bioinformatic analysis of mammalian SWI/SNF complexes identifies extensive roles in human malignancy. Nat. Genet. 45, 592–601 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. 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  CAS  Google Scholar 

  8. Kosho, T. & Okamoto, N. Genotype–phenotype correlation of Coffin–Siris syndrome caused by mutations in SMARCB1, SMARCA4, SMARCE1, and ARID1A. Am. J. Med. Genet. C. Semin. Med. Genet. 166C, 262–275 (2014).

    Article  PubMed  CAS  Google Scholar 

  9. Santen, G.W. et al. Mutations in SWI/SNF chromatin remodeling complex gene ARID1B cause Coffin–Siris syndrome. Nat. Genet. 44, 379–380 (2012).

    Article  CAS  PubMed  Google Scholar 

  10. Tsurusaki, Y. et al. Mutations affecting components of the SWI/SNF complex cause Coffin–Siris syndrome. Nat. Genet. 44, 376–378 (2012).

    Article  CAS  PubMed  Google Scholar 

  11. Deciphering Developmental Disorders Study. Large-scale discovery of novel genetic causes of developmental disorders. Nature 519, 223–228 (2014).

  12. 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 

  13. 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 

  14. Tkachuk, D.C., Kohler, S. & Cleary, M.L. Involvement of a homolog of Drosophila Trithorax by 11q23 chromosomal translocations in acute leukemias. Cell 71, 691–700 (1992).

    Article  CAS  PubMed  Google Scholar 

  15. Kim, K.H. & Roberts, C.W. Targeting EZH2 in cancer. Nat. Med. 22, 128–134 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Varambally, S. et al. The Polycomb group protein EZH2 is involved in progression of prostate cancer. Nature 419, 624–629 (2002).

    Article  CAS  PubMed  Google Scholar 

  17. Lee, W. et al. PRC2 is recurrently inactivated through EED or SUZ12 loss in malignant peripheral nerve sheath tumors. Nat. Genet. 46, 1227–1232 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wu, J.I., Lessard, J. & Crabtree, G.R. Understanding the words of chromatin regulation. Cell 136, 200–206 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Wu, J.I. et al. Regulation of dendritic development by neuron-specific chromatin remodeling complexes. Neuron 56, 94–108 (2007).

    Article  CAS  PubMed  Google Scholar 

  20. Versteege, I. et al. Truncating mutations of hSNF5/INI1 in aggressive paediatric cancer. Nature 394, 203–206 (1998).

    Article  CAS  PubMed  Google Scholar 

  21. Wilson, B.G. et al. Epigenetic antagonism between Polycomb and SWI/SNF complexes during oncogenic transformation. Cancer Cell 18, 316–328 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kia, S.K., Gorski, M.M., Giannakopoulos, S. & Verrijzer, C.P. SWI/SNF mediates Polycomb eviction and epigenetic reprogramming of the INK4b-ARF-INK4a locus. Mol. Cell. Biol. 28, 3457–3464 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. 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 

  24. Li, G. et al. Jarid2 and PRC2, partners in regulating gene expression. Genes Dev. 24, 368–380 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. van der Stoop, P. et al. Ubiquitin E3 ligase Ring1b/Rnf2 of Polycomb repressive complex 1 contributes to stable maintenance of mouse embryonic stem cells. PLoS One 3, e2235 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Feldman, N. et al. G9a-mediated irreversible epigenetic inactivation of Oct-3/4 during early embryogenesis. Nat. Cell Biol. 8, 188–194 (2006).

    Article  CAS  PubMed  Google Scholar 

  27. Ho, L. et al. An embryonic stem cell chromatin remodeling complex, esBAF, is an essential component of the core pluripotency transcriptional network. Proc. Natl. Acad. Sci. USA 106, 5187–5191 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ho, L. et al. An embryonic stem cell chromatin remodeling complex, esBAF, is essential for embryonic stem cell self-renewal and pluripotency. Proc. Natl. Acad. Sci. USA 106, 5181–5186 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Young, R.A. Control of the embryonic stem cell state. Cell 144, 940–954 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Hathaway, N.A. et al. Dynamics and memory of heterochromatin in living cells. Cell 149, 1447–1460 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).

    Article  CAS  PubMed  Google Scholar 

  32. Crabtree, G.R. & Olson, E.N. NFAT signaling: choreographing the social lives of cells. Cell 109 (Suppl.), S67–S79 (2002).

    Article  CAS  PubMed  Google Scholar 

  33. Crabtree, G.R. & Schreiber, S.L. Three-part inventions: intracellular signaling and induced proximity. Trends Biochem. Sci. 21, 418–422 (1996).

    Article  CAS  PubMed  Google Scholar 

  34. Spencer, D.M., Wandless, T.J., Schreiber, S.L. & Crabtree, G.R. Controlling signal transduction with synthetic ligands. Science 262, 1019–1024 (1993).

    Article  CAS  PubMed  Google Scholar 

  35. de Bruijn, D.R. et al. Targeted disruption of the synovial sarcoma–associated SS18 gene causes early embryonic lethality and affects PPARBP expression. Hum. Mol. Genet. 15, 2936–2944 (2006).

    Article  CAS  PubMed  Google Scholar 

  36. Banaszynski, L.A., Liu, C.W. & Wandless, T.J. Characterization of the FKBP.rapamycin.FRB ternary complex. J. Am. Chem. Soc. 127, 4715–4721 (2005).

    Article  CAS  PubMed  Google Scholar 

  37. Schuettengruber, B., Chourrout, D., Vervoort, M., Leblanc, B. & Cavalli, G. Genome regulation by Polycomb and Trithorax proteins. Cell 128, 735–745 (2007).

    Article  CAS  PubMed  Google Scholar 

  38. Blackledge, N.P. et al. Variant PRC1 complex–dependent H2A ubiquitylation drives PRC2 recruitment and Polycomb domain formation. Cell 157, 1445–1459 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. 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  CAS  Google Scholar 

  40. Yen, K., Vinayachandran, V., Batta, K., Koerber, R.T. & Pugh, B.F. Genome-wide nucleosome specificity and directionality of chromatin remodelers. Cell 149, 1461–1473 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Lorch, Y., Zhang, M. & Kornberg, R.D. Histone octamer transfer by a chromatin-remodeling complex. Cell 96, 389–392 (1999).

    Article  CAS  PubMed  Google Scholar 

  42. Mendenhall, E.M. et al. GC-rich sequence elements recruit PRC2 in mammalian ES cells. PLoS Genet. 6, e1001244 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Stanton, B.Z. et al. Smarca4 ATPase mutations disrupt direct eviction of PRC1 from chromatin. Nat. Genet. http://dx.doi.org/10.1038/ng.3735 (2016).

  44. Deal, R.B., Henikoff, J.G. & Henikoff, S. Genome-wide kinetics of nucleosome turnover determined by metabolic labeling of histones. Science 328, 1161–1164 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Buenrostro, J.D., Giresi, P.G., Zaba, L.C., Chang, H.Y. & Greenleaf, W.J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Jadhav, U. et al. Acquired tissue-specific promoter bivalency is a basis for PRC2 necessity in adult cells. Cell 165, 1389–1400 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Schnitzler, G., Sif, S. & Kingston, R.E. Human SWI/SNF interconverts a nucleosome between its base state and a stable remodeled state. Cell 94, 17–27 (1998).

    Article  CAS  PubMed  Google Scholar 

  48. 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 

  49. Khavari, P.A., Peterson, C.L., Tamkun, J.W., Mendel, D.B. & Crabtree, G.R. BRG1 contains a conserved domain of the SWI2/SNF2 family necessary for normal mitotic growth and transcription. Nature 366, 170–174 (1993).

    Article  CAS  PubMed  Google Scholar 

  50. Narayanan, R. et al. Loss of BAF (mSWI/SNF) complexes causes global transcriptional and chromatin state changes in forebrain development. Cell Rep. 13, 1842–1854 (2015).

    Article  CAS  PubMed  Google Scholar 

  51. Yoo, A.S., Staahl, B.T., Chen, L. & Crabtree, G.R. MicroRNA-mediated switching of chromatin-remodelling complexes in neural development. Nature 460, 642–646 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Yoo, A.S. et al. MicroRNA-mediated conversion of human fibroblasts to neurons. Nature 476, 228–231 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Victor, M.B. et al. Generation of human striatal neurons by microRNA-dependent direct conversion of fibroblasts. Neuron 84, 311–323 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Singhal, N. et al. Chromatin-remodeling components of the BAF complex facilitate reprogramming. Cell 141, 943–955 (2010).

    Article  CAS  PubMed  Google Scholar 

  55. Lickert, H. et al. Baf60c is essential for function of BAF chromatin remodelling complexes in heart development. Nature 432, 107–112 (2004).

    Article  CAS  PubMed  Google Scholar 

  56. Tea, J.S. & Luo, L. The chromatin remodeling factor Bap55 functions through the TIP60 complex to regulate olfactory projection neuron dendrite targeting. Neural Dev. 6, 5 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Weinberg, P., Flames, N., Sawa, H., Garriga, G. & Hobert, O. The SWI/SNF chromatin remodeling complex selectively affects multiple aspects of serotonergic neuron differentiation. Genetics 194, 189–198 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Dawson, M.A. & Kouzarides, T. Cancer epigenetics: from mechanism to therapy. Cell 150, 12–27 (2012).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to dedicate this paper to Joe Calarco, who died tragically during the course of this work. His energy, enthusiasm, and insight will continue to guide our work and his warmth brightens our memories.

We are grateful to N. Hathaway, O. Bell, L. Chen, and J. Ronan, for insightful comments and technical assistance. We thank M. Alhamadsheh for synthesis of FK1012. We also thank J. Buenrostro and B. Wu for helpful advice and technical assistance with accessibility studies and V. Petkova. C.K. was supported by the NIH Director's New Innovator Award DP2 (1DP2CA195762-01), the American Cancer Society Scholar Award (RSG-14-051-01-DMC), the Pew Scholar Award, the A.P. Giannini Foundation, the Alex's Lemonade Stand Foundation (ALSF) Young Investigator Award, and the NIH SARC Sarcoma SPORE Career Development and SPORE Project (5U54 CA168512-04) awards. G.R.C. is supported by NIH grants (CA163915, NS046789), CIRM RB4-05886, SFARI and the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Contributions

C.K. and G.R.C. conceived of the study and wrote the paper. C.K., R.T.W., J.P.C., C.M.W., E.L.M., S.M.G.B., and E.J.C. planned and performed the experiments. J.L.P. performed data analysis and statistics.

Corresponding authors

Correspondence to Cigall Kadoch or Gerald R Crabtree.

Ethics declarations

Competing interests

C.K. and G.R.C. are scientific co-founders, shareholders, and consultants of Foghorn Therapeutics, Inc.

Integrated supplementary information

Supplementary Figure 1 Generation of a rapamycin-inducible recruitment system for mSWI/SNF (BAF) complexes to the Oct4 locus in MEFs.

(a) ChIP-seq tracks for Brg1 and Ezh2 over the Oct4 locus in ES cells and MEFs. (b) Lentiviral delivery vector design for Frb-V5-[BAF complex subunit, here shown, SS18] and direct fusion of FKBP to ZFHD1. (ZFHD1-FKBP) for tethering to binding array upstream of the modified Oct4 (Pou5f1) allele. (c) Enrichment of each mark over Oct4:WT and Oct4:CiA alleles in MEFs. The inserted allele and the wild-type allele show quantitatively similar chromatin landscapes. Error bars = Mean ± SD for n=3 experiments. (d) Baf47 and Baf57 Frb-V5 tagged complex subunits properly assemble into BAF complexes. (e) Density sedimentation analysis using 10-30% glycerol gradients indicate that introduced Frb-V5-Ss18 is stably incorporated as endogenous SS18 and is dedicated to the 2MDa BAF complexes. (f) Each Frb-V5 tagged BAF complex can be recruited by 20-40 fold upon 24 hours of rapamycin treatment. Error bars = Mean ± SD for n=3 experiments. (g) BAF complex recruitment (t=30’) to the Oct4:CiA locus in MEFs results in BAF complex occupancy levels similar to those in ES cells. Error bars = Mean ± SD for n=3 experiments. (h) BAF complex recruitment (fold enrichment by ChIP- qPCR) within the recruitment region at (left) +237bp, and (right)+377bp, from the ZHFD1 locus (-309bp and -169bp from the Oct4 TSS) using three different immunocapture antibodies.

Supplementary Figure 2 Chromatin marks, DNA accessibility, gene expression in BAF complex recruitment time course experiments.

(a) (left) Occupancy of PRC2 complexes and H3K27me3 is reduced at 60 minutes post-rapamycin treatment. (right) Recruitment of Frb-V5-Stop does not recruit BAF complexes, nor alter the occupancy of PRC2 or the H3K27me3 mark at t=60 minutes of rapamycin treatment. Error bars = Mean ± SD for n=3 experiments. (b) Total H3, H3K9me3, and H2A.Z are unchanged upon 60- minute BAF complex recruitment. Error bars = Mean ± SD for n=3 experiments. (c) Recruitment of Frb-V5-SS18 (BAF) to the Ascl1 locus. CATCH-IT results indicate nucleosomal turnover at 60’ post treatment with rapamycin. ChIP using V5, H3, H2AK119Ub shows site-specific decreases in histone mark enrichment upon BAF complex recruitment. Error bars = Mean ± SD for n=2 experiments for ChIP and n=4 experiments for CATCH-IT. (d) Schematic for modified ATAC-seq accessibility assays using Tn5 transposase. (e) Tn5 accessibility landscape over the CiA Oct4 locus shows enhanced accessibility within 60 minutes of BAF complex recruitment. (f) Overlap of Brg, Ezh2, and Ring1b MACS-called peaks displayed as Venn diagrams (published datasets). (g) Rapamycin addition (BAF complex recruitment) at various time points indicated does not result in increased percentage of GFP-positive cells (left) nor Oct4 gene expression (right) in CiA Oct4 MEFs. Error bars = Mean ± SD for n=3 experiments.

(h) RNA Pol II ChIP-qPCR over the CiA Oct4 locus indicates lack of recruitment upon BAF complex recruitment over a time course. Error bars = Mean ± SD for n=2 experiments.

Supplementary Figure 3 Structures and downstream reformation of heterochromatin in FK1012 competition (washout) experiments.

(A) Structures of (left) Rapamycin and (right) FK1012 showing the regions that bind FKBP, but not FRB, making FK1012 an effective competitor for rapamycin-induced recruitment of BAF complexes. (B) FK1012 competition (washout) triggers downstream removal of recruited BAF complexes at the +237bp from ZFHD1 site (-309bp from Oct4 TSS). (C-D) Reformation of downstream (C) PRC2 and (D) PRC1 repression upon FK1012 washout at the +237bp (from ZNFHD1 array) site. (E) DNA accessibility is lost and heterochromatin reforms downstream upon FK1012 washout. All experiments are a representative n=1 except Ring1b where error bars = Mean ± SD for n=3 experiments.

Supplementary Figure 4 SS18-SSX- containing (SS-like) BAF complexes display enhanced occupancy and PcG displacement at the Oct4 locus in MEFs.

(A) Recruitment of BAF complexes is similar at the recruitment site (+0bp; -443bp of the Oct4 TSS). (B-D) Eviction of (B) Ezh2, (C) Ring1b, and (D) H3K27me3 at the recruitment site (+0) is similar between WT BAF complexes and SS-like BAF complexes. (E) SS-like BAF complexes show enhanced occupancy downstream (+1034bp) of recruitment site. (F-H) Eviction of (F) Ezh2, (G) Ring1b, and (H) H3K27me3 downstream (+1034bp) of recruitment site is enhanced by SS-like BAF complexes as compared to wild type. Error bars represent Mean ± SEM for n=3 experiments.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–4 and Supplementary Tables 1 and 2 (PDF 1231 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kadoch, C., Williams, R., Calarco, J. et al. Dynamics of BAF–Polycomb complex opposition on heterochromatin in normal and oncogenic states. Nat Genet 49, 213–222 (2017). https://doi.org/10.1038/ng.3734

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3734

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer